Close
About
FAQ
Home
Collections
Login
USC Login
Register
0
Selected
Invert selection
Deselect all
Deselect all
Click here to refresh results
Click here to refresh results
USC
/
Digital Library
/
University of Southern California Dissertations and Theses
/
Trans-epithelial transport of G -CSF -transferrin conjugate
(USC Thesis Other)
Trans-epithelial transport of G -CSF -transferrin conjugate
PDF
Download
Share
Open document
Flip pages
Contact Us
Contact Us
Copy asset link
Request this asset
Transcript (if available)
Content
TRANS-EPITHELIAL TRANSPORT OF G-CSF-TRANSFERRIN
CONJUGATE
by
Adam Scott Widera
A Dissertation Presented to the
FACULTY OF THE GRADUATE SCHOOL
UNIVERSITY OF SOUTHERN CALIFORNIA
In Partial Fulfillment of the
Requirements for the Degree
DOCTOR OF PHILOSOPHY
(PHARMACEUTICAL SCIENCES)
May 2004
Copyright 2004 Adam Scott Widera
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
UMI Number: 3140572
INFORMATION TO USERS
The quality of this reproduction is dependent upon the quality of the copy
submitted. Broken or indistinct print, colored or poor quality illustrations and
photographs, print bleed-through, substandard margins, and improper
alignment can adversely affect reproduction.
In the unlikely event that the author did not send a complete manuscript
and there are missing pages, these will be noted. Also, if unauthorized
copyright material had to be removed, a note will indicate the deletion.
UMI
UMI Microform 3140572
Copyright 2004 by ProQuest Information and Learning Company.
All rights reserved. This microform edition is protected against
unauthorized copying under Title 17, United States Code.
ProQuest Information and Learning Company
300 North Zeeb Road
P.O. Box 1346
Ann Arbor, Ml 48106-1346
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
DEDICATION
This work is dedicated to my wife Linda, and my son Ethan, who make life worth
living. I owe it all to them.
I thank my parents. Their undying support made this work possible....
1 1
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
ACKNOWLEDGEMENTS
First and foremost I thank my thesis advisor, Wei-Chaing Shen, Ph.D., for his
invaluable support and guidance throughout my doctoral studies
I also acknowledge the support offered by the good folks at the Will Rogers Institute
Pulmonary Research Center o f the USC Keck School o f Medicine. Their faithful
collaboration was instrumental to my success. Special gratitude is offered to
Kwang-Jin Kim, Ph.D., whose close reading o f manuscripts provided me with much
excellent teaching and scientific insight.
I thank the American Foundation for Pharmaceutical Education fo r providing
financial support throughout my graduate studies
I thank David Ann, Ph.D., for providing generous use o f his lab’ s equipment and
supplies
Last, but not least, I thank my labmates including, Karin, Jennica, Maureen, Rana,
Rita, Tin-ten, Wendy, Yun Bai, Li Yun, Jeff, Cindy, Wang Li, Fariba, and Thuy for
making life interesting at USC.
Ill
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
TABLE OF CONTENTS
DEDICATION ii
ACKNOWLEDGMENTS iii
LIST OF FIGURES xi
ABSTRACT xv
1. INTRODUCTION 1
1.2. Transferrin, transferrin-receptor, and associated proteins 1
1.2.1. Structure and function of transferrin 2
1.2.2 Transferrin-receptor 6
1.2.2.1 Structure and function of Transferrin-Receptor 6
1.2.2.2 Regulation of expression and tissue distribution 13
1.2.3 Proteins associated with transferrin and transferrin-receptor 15
1.2.3.1. HFE 15
IV
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.2.3.2. Cubilin and Megalin 17
1.3. The recycling and sorting of transferrin receptor 21
1.3.1 Rab proteins involved in transferrin trafficking 22
1.3.2 The Rabl 1 interacting proteins 25
1.3.3 Rab5 effectors, EEAl and PI(3)K, involved in early endosome 25
fusion
1.4. Utilization of TfR for drug delivery 28
1.4.1 TfR-based targeting for anti-cancer therapeutics 28
1.4.2 TfR-mediated transepithelial protein-drug delivery 31
1.4.2.1 anti-TfR antibody drug conjugates for transepithelial drug delivery 32
1.4.2.2 Enhancement of TfR-mediated drug delivery 35
1.4.2.2.1 Lysosomotrophic Amines 36
1.4.2.2.2 Lipophilic carboxylic ionophores 36
1.4.2.2.3 Brefeldin-A 37
1.4.3. Modification of Rab-dependent sorting pathways for enhancement 39
of TfR-mediated gastrointestinal transepithelial transport
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.4.4 Enhancement of TfR-mediated transport in pulmonary epithelium. 42
1. MATERIALS ANB METHODS 48
2.1 Caco-2 Cell Culture 48
2.2 MDCK Cell Culture 48
2.3 Primary Culture of Rat Alveolar Epithelial Cell Monolayers 49
2.4 Measurement of Cell Surface Transferrin Receptor on Caco-2 50
Monolayers
2.5 Measurement of Cell Surface Transferrin Receptor in Rat Alveolar 51
Epithelial Cell Monolayers
2.6 Measurement of Total Cellular Transferrin Receptor in Rat Alveolar 52
Epithelial Cell Monolayers
2.7 TfR-Dependent Uptake of ^^Fe in Caco-2 and Rat Alveolar Epithelial 52
Cell Monolayers
2.8 Immunoflouresence Studies in Rat Alveolar Epithelial Cell 53
Monolayers
2.9 Synthesis of Insulin-Transferrin Conjugate 54
2.10 Cloning and production of G-CSF 56
2.11 Preparation of Tf-G-CSF Conjugate 57
vi
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2.12 Assessment of Apical-to-Basolateral Transcytosis of '^^I-Tf, 60
^^^I-M-Tf, *^^I-G-CSF and ^^^I-G-CSF-Tf across Caco-2 and
MDCK Cell Monolayers
2.13 Assessment of Apical-to-Basolateral Transcytosis of '^^I-Tf, 61
’^^I-GCSF and ‘^^I-Tf-GCSF across Rat Alveolar Epithelial
Cell Monolayers
2.14 Analysis of Transcytosed Proteins 62
2.15 In vivo studies of subcutaneously and orally delivered G-CSF-Tf 63
conjugate in male BDFl mice
2.16 Statistical analyses 64
3. RESULTS 65
3.1 Synthesis and characterization of G-CSF-Tf 65
3.2 The Effect of Wortmannin on TfR-Mediated Transcytosis in 71
Caco-2 Monolayers
3.3 LY294002 Mediated Enhancement of TfR-mediated Transcytosis 74
in Caco-2 Monolayers
3.4 Effect of BFA, AG-10, and LY294002 on TfR-Specific ^^^I-Tf 78
Binding in Caco-2 Monolayers
3.5 Effect of BFA, AG-10, and LY294002 on TfR-specific ^^Fe-Tf 81
uptake in Caco-2 monolayers
V ll
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.6 Comparison of Transcytosis Enhancers’ Effect on TfR-Mediated 84
Transport in Caco-2 Monolayers
3.7 The effect of LY294002 on TfR-mediated transport of a G-CSF-Tf 87
conjugate across Caco-2 monolayers
3.8 Comparison of the effect of BFA, AG-10, or LY294002 treatment 90
on TfR-mediated Transcytosis in MDCK monolayers
3.9 Effect of BFA, AG-10, and LY294002 on TfR-Specific ‘^^I-Tf . 94
Binding in MDCK Monolayers
3.10 Effect of BFA, AG-10, and LY294002 on TfR-Specific *^^I-Tf 97
Binding in MDCK Monolayers
3.11 Measurement of total surface transferrin receptor in primary cultures 100
of rat alveolar epithelial cell monolayers
3.12 Total cellular TfR in rat alveolar epithelial cell monolayers as a 102
function of culture day
3.13 Rat alveolar epithelial cell monolayers exposed to keratinocyte 103
growth factor during culture retain TfR expression
3.14 Confocal microscopy of keratinocyte growth factor treated rat 103
alveolar epithelial cell monolayers
3.15 Type II cell-like rat alveolar epithelial cell monolayers exhibit 107
polarized distribution of TfR at the cell surface
3.16 Type II cell-like rat alveolar epithelial cell monolayers exhibit 108
polarized TfR-specific ^^Fe-Tf uptake
via
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.17 Effect of BFA, AG-10, and LY294002 on TEER of type II cell-like 109
rat alveolar epithelial cell monolayers
3.18 Effects of BFA on distribution and endocytosis of TfR in rat 112
alveolar epithelial cell monolayers
3.19 TfR-mediated apical-to-basolateral Tf transport in BFA-treated 117
type n cell-like rat alveolar epithelial cell monolayers
3.20 TfR-mediated apical-to-basolateral transcytosis of In-Tf conjugate 120
across type II cell-like rat alveolar epithelial cell monolayers
3.21 Analysis of ^^^I-In-Tf transcytosed across type II cell-like rat 122
alveolar epithelial cell monolayers
3.22 TfR-Mediated ^^^I-GCSF-Tf transcytosis in type II cell-like rat 122
alveolar epithelial cell monolayers
3.23 Comparison of ^^^I-GCSF-Tf and ^^^I-GCSF apical-to-basolateral 125
transport in AEG monolayers
3.24 Analysis of transcytosed ^^^I-GCSF-Tf 126
3.25 Apical-to-basolateral Transcytosis of G-CSF-Tf and TF across 129
Caco-2 monolayers
3.26 Chromatographic analysis of transcytosed G-CSF-Tf 131
3.27 Analysis of biological activity of transcytosed '^^I-G-CSF-Tf 131
in Caco-2 monolayers
IX
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.28 Neutrophil proliferation in BDFl mice dosed with subcutaneous 134
and oral G-CSF-Tf
4. DISCUSSION 139
5. CONCLUSIONS 165
6. FUTURE DIRECTIONS 170
7. ALPHABETIZED BIBLIOGRAPHY 174
8. APPENDIX: Numerically Ordered References 216
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
LIST OF TABLES A N D FIGURES
Table 1) Total Cellular TfR 102
Figure 1) The classical endocytosis pathway of transferrin-receptor
(TfR).
Figure 2) The x-ray crystal structure of the transferrin receptor
ectodomain.
Figure 3) The sequence and secondary structure of human transferrin 10
receptor.
Figure 4) Synthesis scheme of In-Tf conjugate. 55
Figure 5) Synthesis scheme for G-CSF-Tf. 59
Figure 6) G-CSF-dependent proliferation of NFS-60 cells. 67
Figure 7) SDS-PAGE analysis of G-CSF-Tf. 69
Figure 8) Chromatographic analysis of G-CSF-Tf. 70
Figure 9) Wortmannin induced enhancement of TfR-mediated 72
transcytosis in Caco-2 monolayers.
Figure 10) Wortmannin’s effect on TEER of Caco-2 monolayers. 73
XI
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 11) LY294002 induced enhancement of TfR-mediated 75
transcytosis in Caco-2 monolayers.
Figure 12) LY294002’s effect on TEER of Caco-2 monolayers. 77
Figure 13) The effect of BFA, AG-10, and LY294002 on the surface 79
distribution of TfR in Caco-2 monolayers.
Figure 14) The effect of BFA, AG-10, and LY294002 on the uptake of 82
^^Fe-Tf in Caco-2 monolayers.
Figure 15) Comparison of transcytosis enhancer effect on TfR-mediated 85
transcytosis in Caco-2 monolayers.
Figure 16) LY294002 induced enhancement of TfR-mediated 88
transcytosis of ^^^I-G-CSF-Tf across Caco-2 monolayers.
Figure 17) Comparison of transcytosis enhancer effect on TfR-mediated 91
transcytosis in MDCK monolayers.
Figure 18) Effect of BFA, AG-10, and LY294002 treatment on TEER 93
of MDCK monolayers.
Figure 19) The effect of BFA, AG-10, and LY294002 on the surface 95
distribution of TfR in MDCK monolayers.
Figure 20) The effect of BFA, AG-10, and LY294002 on the uptake of 98
^^Fe-Tf in MDCK monolayers.
Figure 21) Total surface TfR in rat alveolar epithelial cells (AEC) as a 101
function of day in culture.
XII
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 22) Effect of KGF treatment on TfR expression in rat alveolar 104
epithelial cell monolayers.
Figure 23) KGF treatment retains TfR expression in rat alveolar 105
epithelial cell monolayers.
Figure 24) Rat AEC monolayers tested on day 7 for polarized 108
distribution of TfR.
Figure 25) TfR-specific uptake of ^^Fe in rat AEC monolayers on day 7. 110
Figure 26) Effect of BFA, AG-10, and LY294002 treatment on TEER 111
of rat type II cell-like alveolar epithelial cell monolayers.
Figure 27) The effect BFA on surface TfR expression and ^®Fe-Tf 114
uptake in rat alveolar epithelial cells (AEC).
Figure 28) The effects of BFA on ^^^I-Tf transport in rat alveolar 118
epithelial cell monolayers.
Figure 29) *^^I-In-Tf transport in rat alveolar epithelial cell monolayers. 121
Figure 30) Specific TfR-mediated transport of ^^^I-GCSF-transferrin 123
conjugate determined on day 7 and comparison to ’^^I-GCSF.
Figure 31) Gel filtration chromatogram of TfR-mediated transcytosed 127
'^^I-GCSF-Tf.
Figure 32) Evaluation of biological activity of transcytosed 128
GCSF-transferrin.
X lll
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 33) Specific TfR-mediated transport of ^^^I-GCSF-transferrin 130
conjugate determined in Caco-2 monolayers and comparison
to ‘^^I-GCSF.
Figure 34) Gel filtration chromatogram of TfR-mediated transcytosed 132
*^^I-GCSF-Tf.
Figure 35) Evaluation of biological activity of transcytosed G-CSF-Tf. 133
Figure 36) Myelopoietic effect of subcutaneously administered 136
G-CSF-Tf.
Figure 37) Myelopoietic effect of orally administered G-CSF-Tf 137
Figure 38) Dose response effect of orally administered G-CSF-Tf 138
Figure 39) Spacefill model of G-CSF x-ray crystal structure. 140
Figure 40) Possible mechanisms of transcytosis enhancer’s effect on 147
TfR distribution in polarized epithelia.
Figure 41) PI3K Inhibitors as potential TfR-mediated transcytosis 148
enhancers.
Figure 42) Possible G-CSF-Tf fusion constructs. 171
XIV
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
ABSTRACT
The purpose of this study was to investigate the transferrin-receptor (TfR)-
mediated transepithelial transport of G-CSF-transferrin (If) conjugate in cultured
enterocyte-like Caco-2 monolayers, rat alveolar epithelial cell monolayers, and the
myelopoietic effect of subcutaneously and orally administered G-CSF-Tf in BDFl
mice.
Methods included using Caco-2 and rat alveolar epithelial cell monolayers
exhibiting a minimum transepithelial electrical resistance of 500 Qcm^ and 2000
Qcm^, respectively as an in vitro model trans-epithelial transport capability. BDFl
mice were used as an in vivo absorption model. TfR-mediated transcytosis was
measured by using '^^I-G-CSF-Tf and analyzing the downstream compartment by
gamma counter. The efficacy of subcutaneously and orally administered G-CSF-Tf
was determined by performing daily absolute neutrophil counts.
The results obtained from transport experiments in Caco-2 cells and rat
alveolar epithelial cell monolayers revealed that the monolayers that received *^^I-G-
CSF-Tf exhibited significantly higher apical-to-basolateral transport rates compared
to the monolayers that received *^^I-G-CSF. Inclusion of 100-fold excess unlabeled
Tf reduced the extent of ^^^I-G-CSF-Tf transport by 80%. Chromatographic and
bioactivity assays revealed that the protein recovered from the basolateral
compartment was the intact conjugate and it retained full ability to stimulate the
proliferation of the G-CSF dependent cell line, NFS-60, upon reduction.
XV
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Subcutaneous administration of G-CSF-Tf in BDFl mice demonstrated that the
conjugate is able to elicit a statistically significant enhancement in therapeutic effect
relative to filgrastim, which includes a longer duration of action with higher absolute
neutrophil counts. Oral administration of G-CSF-Tf in BDFl mice demonstrated
that G-CSF-Tf is able to elicit a significant, and apparently dose dependent, increase
in absolute neutrophil counts while filgrastim had no effect.
Our data indicate that G-CSF-Tf is transported across Caco-2 monolayers and
ratalveolar epithelial cell monolayers by TfR-specific processes, at a rate that is
significantly higher than the non-specific flux of G-CSF. G-CSF-Tf is also able to
elicit a prolonged myelopoietic effect relative to filgrastim when administered by
subcutaneously or orally in BDFl mice. The development of an orally or pulmonary
bioavailable G-CSF has the potential to provide great benefit to patients under
sustained G-CSF dosing regimes.
XVI
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1. INTRODUCTION
Iron is a fundamental necessity for almost every living organism. The ability
of this metal to readily exist in two common redox states leads to its requirement as a
co-factor in many fundamental biological processes, including O2 absorption, ATP
generation through oxidative phosphorylation, DNA synthesis, etc. However, the
charged and reactive nature of free iron presents a challenge for efficient tissue
distribution and cellular uptake. Transferrin (Tf) and its cognate receptor have been
used by vertebrate species to overcome these barriers for efficient and regulated
intracellular iron delivery. The ubiquitous nature of the TfR and the fact that it is
internalized through endocytic means has made it an essential tool for the general
elucidation of sorting and recycling pathways of endocyticaUy derived vesicles in
epithelial cells. In addition, the high specificity of endocytic uptake of Tf by TfR has
made it a subject of interest for targeted drug delivery via parenteral administration
and for delivery of TfR-targeted drug conjugates across epithelial barriers.
The introduction of this work focuses on the sorting and recycling pathways
of TfR in epithelial cell systems. Special focus is given to the methods by which the
understanding of these pathways can be exploited to achieve higher efficiency in
targeted delivery systems.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.2 Transferrin, transferrin-receptor, and associated proteins
1.2.1 Structure and function of transferrin
Transferrins are a structurally related class of metal-binding glycoproteins of
approximately 80 kDa in size whose primary function is the binding and
transportation of non-heme iron (1-5). The transferrins have been classified into
three major sub-classes: serum Tf, lactoferrin, and ovotransferrin. Serum Tf is
responsible for binding and transporting iron through many of the biological fluids
including blood, lymph, cerebrospinal fluid, colostrum, bile, amniotic fluid, and
breast milk (6-10). Lactoferrin is found in breast milk, saliva, and tear secretions; it
is thought to function primarily in these environments as an iron chelator,
sequestering iron to act as a bacteriostatic agent (11-13). Lactoferrin also is
implicated in modulating leukocyte antibacterial activity and may also be involved
with inflammatory responses (14; 15). Unlike serum Tf, lactoferrin does not
contribute iron to erythrocytes, and it is cleared by the reticulo-endothelial system
(16). Ovotransferrin is found in reptile and avian oviduct secretions as well as avian
egg white, and its function is analogous to that of lactoferrin, purported to primarily
serve as a bacteriostat via the sequestration of iron (17-19). In addition to the three
major types of Tf, melanotransferrin (p97) can also be considered to be part of the Tf
family. This fourth member of the Tf family is a structural homologue distinctly
different from the others in that it seems to have little to do with the receptor-specific
cellular uptake of iron (20; 21). Rather, this protein is predominantly localized to the
surface of human melanomas and may assist in rapid cell prohferation via iron
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
scavenging and the prevention of lipid peroxidation (22; 23). Considering that serum
Tf has been used extensively in endocytosis research, and since it is the protein that
has been identified as a potential drug carrier candidate, it will be the focus of the
remainder of the review; the other three transferrins will not be discussed further.
The serum transferrins from different species share a high degree of
homology, typically composed of a single chain polypeptide with approximately 700
amino acids. However, there are varying degrees of carbohydrate content between
species (24). Human Tf is glycosylated with identically branched heterosaccharide
chains linked to the polypeptide by aspariginyl-iV-acetylglucosamine bonds at two
separate residues (25) and one O-linked carbohydrate at Ser51 . While the
carbohydrate chains of Tf are seen in all species, they do not affect the ability of Tf
to bind to TfR, nor do they affect they ability of the receptor complex to be
internalized into the cell (26; 27). In addition, in contrast to other proteins, the
desialation of Tf has little effect on the catabolism rate (28).
Transferrin is composed of two homologous globular domains, known as the
N and C lobes, connected by a short Unear peptide region, that function in a similar
fashion. Both lobes of the Tf molecule are capable of binding one iron atom with
similar affinities. However, one of the necessities for stringent binding of iron to Tf
is the presence of synergistic bicarbonate anions (29-31), which allows F e^"^ to
tightly coordinate with the amino acids that are suspected of comprising the iron
binding pocket of the N and C lobes (i.e., two Tyr, one Lys and one His for each
lobe) (32; 33). Lack of the presence of bicarbonate anion also affects the abiUty of
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Tf to properly release Fe^^ once it is bound, with bicarbonate-free Tf demonstrating
poor iron release capabilities (34; 35). In addition, Tf has been suggested as a
suitable carrier for medically therapeutic metal ions, since Tf has the ability to bind
to other metal ions in vitro (36-39). However there is little evidence to suggest that
Tf has significant interactions with metals other than iron under normal in vivo
conditions (7).
One of the unique features of Tf is that the binding of Fe^^ alters the shape of
the molecule, affecting the binding affinity of Tf for its receptor. This
conformational change seems to occur in a stepwise fashion as each iron molecule is
bound (40-42). The iron-loaded Tf, or holotransferrin, binds to the receptor with
greater affinity than mono-transferrin or apo-transferrin (43; 44). Binding of iron
also results in increased stability of the molecule relative to the apo-form, with the
iron-loaded molecule exhibiting increased resistance to thermal or proteolytic
degradation (45; 46).
Transferrin undergoes a conformational change as a result of a trigger-
mechanism that is induced by the decrease in pH during the endocytosis cycle. This
allows for the release of iron into the endosomal compartment of the cell and the
subsequent recycling of the apo-transferrin to the cell surface, where the lowered
affinity of the apo-transferrin for the receptor at extracellular pH causes the release
of Tf from the receptor (47; 48). A schematic of the endocytosis and recycling cycle
for the Tf/TfR complex is shown in Figure 1.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(5)
©
Clathrln-
coated pit
Figure 1: The classical endocytosis pathway of transferrin-
receptor (TfR). Transferrin (Tf) binds two atoms per molecule.
The iron-loaded Tf is subsequently bound by TfR and the Tf/TfR
complex is internalized via clathrin coated pits. The iron is released
from Tf in the acidic endosomal environment and Tf/TfR is recycled
back to the plasma membrane. Iron-free Tf is then released from TfR
to complete the cycle.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
The liver is the principle site of Tf production and catabolism. This can be most
dramatically demonstrated in the case of human liver transplant patients, who after
receiving a new liver, obtain the electrophoretic profile of the donor’s serum Tf (as
determined by the normal inter-individual variability in post-translational
modification of Tf) (49; 50). In addition to this example, in vitro studies with rat
liver have determined that the rates of incorporation of radiolabeled amino-acids into
transferrin-precursors were nearly identical to the total synthesis values obtained by
in vivo perfusion studies (51-53). While the liver is responsible for the large
majority of Tf catabolism, a significant amount of Tf is also secreted by the liver into
the bile, with estimates for the average normal adult of approximately 100 mg/day
passing into the small intestine (54).
1.2.2 Transferrin-receptor
1.2.2.1 Structure and function
TfR was first recognized through formation of monoclonal antibodies raised from
mice that were immunized against various neoplastic cell lines, with several of the
antibodies recognizing antigenic portions of the protein we now know as TfRl (55-
57). The receptor is a homodimer composed of two identical subunit monomers with
an approximate mass of 90 kDa each (58). The ectodomain of the receptor is
composed of three distinct domains, the protease-like, apical, and helical domains
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(Figure 2). The helical domains of TfR face each other and interact to form the
dimeric receptor (59). The extracellular portion of the receptor is subject to extensive
post-translational modification with three sites of mannose rich iV-linked
glycosylation at residues Asn251, Asn317, and Asn727 and one O-linked
glycosylation on Thrl04 (60). The monomers are covalently linked by two disulfide
bonds at residues Cys 89 and Cys98 (61). Treatment with trypsin cleaves two 70
kDa fragments that retain the ability to bind to Tf. The protein also has a single
transmembrane domain, comprised of residues 68-88 of the polypeptide. Following
this, it has a short cytoplasmic tail, of 5 kDa in size, that is the site of
phosphorylation and acylation of the receptor (61). Unlike Tf, the post-translational
modifications of TfR seem to be crucial for its biological functionality. Specifically,
mutations that eliminate the sites of potential A-linked glycosylation severely limits
the ability of the receptor to bind to Tf.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Apical
Domain
Domaifi
Figure 2: The x-ray crystal structure of the transferrin receptor
ectodomain. The putative dimeric orientation of two transferrin
receptor molecules is shown as suggested by Lawrence et al. The
receptor is organized into three distinct domains, composed of the
protease-like, apical, and helical domains. The stalk region is also
shown with the suggested orientation relative to the protease-like
domain and the plasma membrane. The image was rendered from
the pdb coordinates of the A subunit of 1CX8 (human TfR,
www.rcsb.org) (Lawrence et al, 1999) using RasMol and Adobe
Photoshop.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
In addition, mutations to the 0-linked glycosylation at Thrl04 suggest that reduction
in this type of carbohydrate linkage may limit the cellular half-life of the receptor
(62; 63). The sequence and secondary structural motifs of TfR is shown in Figure 3.
The receptor has very high affinity for differic-Tf, with binding constants on
the order of 2-7 x 10'® M (64). The exact orientation of Tf binding to TfR is not fully
understood, however the primary recognition site appears to reside on the C lobe of
Tf, with a sequential synergistic effect between the N and C lobes of Tf that
enhances affinity for TfR (65; 66). In addition, site-directed mutagenesis studies
have shown that the central helical domains of TfR, formed by the union of the two
monomers, are key to the ability of the receptor to recognize Tf (67). However,
inspection of the computational union of Tf and the TfR crystal structures
(ectodomain) have also indicated that much favorable interaction occur between the
outward facing apical domains of the receptor and Tf (59). The iron saturation status
of Tf also plays a large role in the ability of TfR to recognize Tf, with apo-Tf having
very little affinity for TfR at physiological pH 7.4 (43). However, at pH 5.0 Tf
forms a very stable complex with TfR, which is the underlying basis for the iron
exchange and transport capability of Tf (68-70).
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 3; The sequence and secondary structure of human transferrin
receptor.
Each residue in the sequence is reported as a single letter code.
Secondary structure is calculated and described according to an
implementation of the method of Kabsch and Sander (1983) Biopolymers
22, 2577-2637. The assignments are: H = helix; B = residue in isolated beta
bridge; E = extended beta strand; G = 310 helix; 1 = pi helix; T = hydrogen
bonded tum; S = bend. From the Protein Data Bank, www.rcsb.org.
10
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1 LYWDDLKRKL SEKLDSTDFT STIKLLNENS YVPREAGSQK DENIALYVEN
HHHHHHHH HHHHHT HH HHHHHTTTGG GSS TTSHH HHHHHHHHHH
51 EFREFKLSKV WRDQHFVKIQ VKDSAQNSVI IVDKNGRLVY LVENPGGYVA
HHHHTT S EEEEEEEEE EE SS EEE EE SSS S B SS BT
101 YSKAATVTGK LVHANFGTKK DFEDLYTPVN GSIVIVRAGK ITFAEKVANA
T EEEEE EEE TTTTTT TTT SSS T TSEEEEES S S HHHHHHHH
151 ESLNAIGVLI YMDQTKFPIV NAELSFFGHA HLGTGDPYTP GFPSFNHTQF
HHTT EEE E TTTS S SS B B SSSS TT S SS SSSS
201 PPSRSSGLPN IPVQTISRAA AEKLFGNMEG DCPSDWKTDS TCRMVTSESK
S SS S S EEEE HHH HHHHHHTB S SSSSS T T SEE TT
251 NVKLTVSNVL KEIKILNIFG VIKGFVEPDH YVWGAQRDA WGPGAAKSGV
EEEEEE EEE EEEEEEEE EE SSBTT EEEEE SS TTTTHH
301 GTALLLKLAQ MFSDMVLKDG FQPSRSIIFA SWSAGDFGSV GATEWLEGYL
HHHHHHHHHH HHHHHHHTT SB EEEE E SGGGTTH HHHHHHGGGT
351 SSLHLKAFTY INLDKAVLGT SNFKVSASPL LYTLIEKTMQ NVKHPVTGQF
TTHHHHB EE EE TT S S S EEEE GG GHHHHHHHHT T B SSSSSB
401 LYQDSNWASK VEKLTLDNAA FPFLAYSGIP AVSFCFCEDT DYPYLGTTMD
SS TT SS B TTTHH HHHHHTS EEEEE SS SSTTBTT
451 TYKELIERIP ELNKVARAAA EVAGQFVIKL THDVELNLDY EEYNSQLLSF
HHHHHTTTT THHHHHHHHH HHHHHHHHHH HHSSS S SHHHHHHHHH
501 VRDLNQYRAD IKEMGLSLQW LYSARGDFFR ATSRLTTDFG NAEKTDRFVM
HHHHHTTHHH HHHSS HH HHHHHHHHHH HHHHHTHHHH HTTTT HHHH
551 KKLNDRVMRV EYHFLSPYVS PKESPFRHVF WGSGSHTLPA LLENLKLRKQ
HHHHHHHHHH TGGGBTTTTT TTT S BTT T STTTHHH HHHHHHHHHT
601 NNGAFNETLF RNQLALATWT IQGAANALSG DVWDIDNEF
SS 3 HHHH HHHHHHHHHH HHHHHHHS S SSS
11
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Within the past few years a new member of the TfR family was discovered
and denoted as transferrin receptor 2 (TfR2). However, unlike TfRl, TfR2 has a
much lower affinity for Tf (25 fold lower than TfRl) and it seems to be primarily
involved with regulation and maintenance of iron homeostasis in the body, and it
might also play a minor role in the delivery of iron to rapidly dividing tissues. The
TfR2 gene produces two transcripts of 2.5 and 2.9 kb in length. Analysis of the
amino acid sequence suggests that TfR2 is a type II transmembrane glycoprotein,
like TfRl, sharing 45% sequence identity and 66% similarity in the extracellular
domain (71). In contrast to TfRl, TfR2 is primarily expressed in the liver (71; 72).
It lacks iron-regulatory elements in the coding and noncoding regions of its mRNA,
indicating that the expression of TfR2 is not regulated by the iron-responsive protein
feedback mechanism, as seen for TfRl. It would appear from this that TfR2 plays a
minor role in regulating intracellular iron concentrations, or it perhaps responds to a
yet undiscovered intracellular regulatory mechanism (73). In spite of these
differences, TfR2 interacts with Tf in a similar fashion as TfRl. Specifically, TfR2
exhibits changes in binding affinity to Tf as a result of environmental pH and the
iron saturation status of Tf. Specifically, TfR2 does not bind very well to apo-Tf at
physiological pH, demonstrating a preference for holo-Tf, while an acidic milieu
results in preferential binding of the apo-Tf (73). While there has been some
indication that TfR-2 expression levels may be a marker of erythroid precursor cells,
as levels of TfR2 mRNA levels in erythroleukemia marrow samples are significantly
elevated (74; 75), most of the data seem to support a role for TfR2 in modulating
12
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
iron homeostasis. For example, mutations in TfR2 have been associated with a form
of hereditary hemochromatosis that is not associated with forms of hemochromatosis
that arise from mutations in the hemochromatosis protein (HFE) (76-79), suggesting
role for TfR2 in excessive iron uptake. In addition, TfR2 has also been implicated in
regulating expression of hepcidin, a modulator of dietary iron uptake (80).
1.2.2.2 Regulation of expression and tissue distribution
TfRl expression is regulated at the post-transcriptional level by interactions between
iron-regulatory proteins (IRP) and iron-response elements (IRE). The IRE are
portions of the 3’-untranslated portion of TfR mRNA that form secondary structural
features in the shape of five hairpins (81; 82). These hairpins are required for the
iron-dependent regulation of TfR expression. The IRE of TfR mRNA are recognized
and bound by the IRP, thus controlling the extent of TfR mRNA translation and
stability (83). Under iron deprivation conditions, IRP bind to the IRE increasing the
stability of the mRNA and thus up-regulating the expression of TfR (84-86). There
are two distinct IRP that interact with TfR IRE, known as IRPl and IRP2. BRPl and
IRP2 respond to variations in iron concentration via differing mechanisms (83; 87).
IRPl has been considered to be a bifunctional enzyme, functioning in a
similar fashion to aconitase when cellular iron levels are plentiful, with no RNA
binding ability, and subsequently switching to a predominantly RNA binding protein
functionality when cellular iron levels are low (88-92). IRPl also shows dual
13
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
functionality with regard to regulation of intracellular iron concentrations. For
example, not only does it bind the IRE of TfR to up-regulate TfR production, but
evidence suggests that in iron-rich conditions it also binds the 5’-region of ferritin
mRNA to inhibit the complete translation of the ferritin protein(93-96). The
resultant down-regulation of ferritin would conceivably result in the complementary
liberation of free iron from intracellular storage.
In contrast, IRF2 synthesis is directly affected by intracellular iron levels,
with regulation occurring at the ubquitination level (97-99). For example, when iron
stores are plentiful, IRP2 rapidly undergoes ubiquitination and is degraded in the
cellular proteosome machinery (97). In conditions where intracellular iron levels are
low, IRP2 is up-regulated by the renewed production of non-proteosome directed
protein (100; 101). While there are differences in the regulation of IRPl and IRP2 in
response to iron levels, their functional significance in relation to TfR is not
necessarily mutually exclusive (102). While constitutive expression of mutant
inactive IRPl in vitro may adversely affect normal iron-metabolism (103), in vivo
data with mice that are IRPl deficient demonstrate no abnormalities in iron
utilization, implying that when intracellular IRPl is lacking, IRP2 may be sufficient
to successfully regulate TfR expression (104).
The regulation of TfR2 is not well understood at the cellular level. For
example, in contrast to TfRl, chelation of iron does not up-regulate the expression of
TfR2 (105). Indeed, in several instances TfR2 behaves in a manner opposite to
TfRl. For example, in liver development, TfR2 is up-regulated, while TfRl is
14
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
down-regulated; conversely, during erythrocytic differentiation of murine
erythroleukemia cells, expression of TfRl steadily increases, while TfR2 expression
is down-regulated (75).
TfR is expressed in many tissue types in the body, particularly those areas
that feature a normally high turnover of cells. The level of receptors is particularly
high in erythrocyte precursors, placenta, and the liver (106-110). Of particular note
is the presence of TfR in appreciable amounts on in the capillaries of the brain (111),
epithelium of the small intestine (112; 113), type II pneumocytes (114), and
neoplastic carcinomas (115). Knowledge of the TfR present in these tissues had led
to the focus of targeting of TfR for the delivery of therapeutic molecules.
1.2.3 Proteins associated with transferrin and transferrin-receptor
I.2.3.1 HFE
Hereditary hemochromatosis is a genetic disorder that affects approximately 1 in 200
Caucasians. It is a disease characterized by excess dietary iron absorption and iron
deposition in several tissues. Clinical consequences include hepatic failure,
hepatocellular carcinoma, diabetes, cardiac failure, impotence, and arthritis.
Although an increase in plasma iron levels may be evident early in life, in most cases
these clinical symptoms do not present themselves until age 40 to 50, as iron-levels
15
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
are slowly but steadily increased throughout the life of the individual (116; 117).
The symptoms of hereditary hemochromatosis are thought to derive from a defect in
the hemochromatosis gene which results in a decrease in the amount of functional
gene product, known as HFE, a protein closely related to the major
histocompatibility complex class I molecules (118). HFE is preferentially expressed
in duodenal crypt cells and Kupffer cells of the liver, predominantly localized to the
perinuclear region of the cell (119; 120). The HFE protein is a 343-residue type I
transmembrane glycoprotein that contains a membrane bound heavy chain (like
MHC class I proteins) with three extra-cellular domains (121). However, unlike the
MHC class proteins, two of the helices in the extra-cellular domain are spaced much
closer together, eliminating what would normally be the peptide-binding groove for
the MHC class I proteins. This difference reflects the fact that HFE is important for
modulating Tf-mediated iron uptake rather than peptide binding (122; 123). The
majority of hemochromatosis patients have a defect in a single codon of the gene that
results in the mutation of Cys282 to Tyr282, the result of which is the abolishment of
a crucial disulfide bond in HFE (124; 125). Shortly after the discovery of the HFE
gene, it was shown that HFE closely interacts with the Tf receptor (121; 122; 126;
127). In addition, confirmation of HFE’s role in hemochromatosis was provided by
gene knockout studies in mice where the HFE knockout mice demonstrated elevated
Tf saturation and increased iron storage in hepatocytes by 10 weeks of age (128).
HFE binds to TfRl with a high affinity at physiological pH of 7.4, similar in
magnitude to Tf, and it competes with Tf for binding to TfR, reducing the affinity of
16
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
the receptor for Tf in a noncompetitive manner (123; 129). Unlike apo-Tf, HFE has
very low binding affinity for TfR at pH 6.0, which is representative of the
environment that would be seen in the early part of the endosomal pathway (121).
HFE is closely related to the TfR-specific iron uptake pathway (130-132), and it co-
localizes with TfR rich intracellular compartments (127) and with TfR on the
basolateral surface of enterocyte precursor cells (120). It has been shown that HFE
binding to TfR is required for HFE to be transported to TfR positive endosomes and
for the regulation of iron homeostasis in cultured cells, but binding is not required for
HFE to be transported to the basolateral membrane (133). While HFE has been
shown to avidly bind to TfRl, it has very little affinity for TfR2 (134). HFE, through
its interactions with Tf and TfR, is implicated in sensing and regulating iron-levels in
the body, particularly at the level of iron uptake through the duodenal enterocyte. It
is possible that HFE is not the sole mechanism by which the iron-body stores are
regulated, as HFE deficient mice are still capable of regulating uptake of iron from
the diet to some extent (128; 135), but HFE plays a vital role in regulating iron-
stores. The exact mechanism of HFE and Tf/TfR interactions that lead to iron uptake
regulation remains to be discovered.
1.2.3.2 Cubilin and Megalin
Cubilin and megalin are large cell-surface receptor proteins that are the mediators of
endocytic uptake for a wide array of ligands. These two proteins, depending upon
17
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
the tissue, are closely associated but structurally very different. Both receptors are
expressed primarily on the apical surfaces of polarized epithelial cells. Megalin and
cubilin bind distinct ligands with variable affinities. In some cases, the two proteins
act independently of one another, while in other situations the two receptors are co
expressed and act in concert with one another as a dual-receptor complex. The
cubilin/megalin receptor system is of interest to this review because recent evidence
has suggested that this receptor system is a major pathway for the uptake of Tf from
the apical membranes of polarized epithelia.
Cubilin is a 3600 amino acid protein with a non-glycosylated molecular
weight of approximately 400 kDa. Post-translational modification of the protein
results in the mature protein with a molecular weight of 460 kDa (136; 137). The
complete cDNA sequence has been elucidated for human (138), canine (139), and rat
(137); it shows a high degree of homology between the species. Analysis of the
sequences revealed that cubilin is composed of an initial region of roughly 110
amino acids followed by eight epidermal growth factor (EGF)-like domains
followed by 27 CUB (complement subcomponents Clr/Cls, EGF-related sea urchin
protein and bone morphogenic protein-1) domains (140). While cubilin is believed
to be bound to the plasma membrane of the cell at its amino terminus (141), no
transmembrane segments have been identified in the protein, and it does not have a
glycosylphosphatidylinositol (GPI)-anchor. Helical plotting of the amino-terminal
region of cubilin has suggested that an amphipathic helix is formed by the protein
that is similar to the lipid binding regions of apolipoproteins (141). It has been
18
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
suggested that cubilin is anchored to the plasma membrane by the insertion of the
hydrophobic residues of the conserved helices into the lipid bilayer, as is postulated
for other proteins (142-146). In addition, cubilin has a potential palmitoylation site at
the amino terminal region which could further assist in anchoring the protein to the
membrane (147; 148).
Megalin is a 4600 amino acid type I transmembrane receptor protein with a
non-glycosylated molecular weight of roughly 517 kDa. Post-translational
modification results in the mature protein with a molecular weight near 600 kDa.
Megalin belongs to the low-density lipoprotein receptor family (149). The complete
cDNA sequences have been elucidated in rat (150)and human (151); there is 77%
identity between the species. The ectodomain of megalin consists of four lipid
binding regions composed of cysteine-rich complement-type/LDLR class A repeats.
These are separated by 17 EGF-like repeats and eight cysteine-poor spacer regions,
which contain YWTD motifs and are involved in the pH-sensitive release of ligands
in endosomal compartments (152). The short carboxy-terminal cytoplasmic tail of
megalin mediates clustering in clathrin coated pits and it contains certain motifs that
have been implicated in the genesis of signal transduction (153; 154).
Megalin and cubilin are co-expressed at the plasma membrane and
throughout the endosomal system of epithelial cells. In particular, they are expressed
in several absorptive epithelia, including the placental cytotrophoblast (155; 156),
visceral yolk sac (157; 158), renal proximal tubule (157; 159-161), and the small
intestine (162; 163). In the kidney proximal tubule and visceral yolk sac, the two
19
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
receptors have been colocalized at the brush border membrane, coated pits, endocytic
vesicles and dense apical tubules, which comprise the membrane-recycling
machinery in these cells (157; 164; 165). In addition, studies have also suggested
that megalin and cubilin have some limited presence in lysosomal compartments.
Cubilin has recently been shown to be a receptor for Tf. In an elegant study
by Kozyraki et a l, the endocytosis of Tf by cubilin was shown to be accomplished in
a megalin-dependant fashion (166). This hypothesis is supported by the finding that
lysosomes of human, dog, and mouse renal proximal tubules strongly accumulate Tf,
whereas no Tf is detectable in the endosomal/lysosomal system of the renal tubule
epithelium of dogs with deficient surface expression of cubilin. Elevated levels of Tf
are seen in the urine of these dogs. Mice with deficient expression of megalin also
excrete excessive levels of Tf in the urine. In addition, it was demonstrated that in
megalin-deficient murine kidney-proximal-tubule epithelial cells, Tf is localized to
the lumenal plasma membrane, but not in endocytic vesicles, indicating defective
internalization of the cubilin/megalin complex. Cubilin may be responsible for a
major portion of the catabolism of Tf in the body. After the liver, the kidney is the
site of the majority of Tf catabolism. In addition, the cubilin-mediated endocytosis
and lysosomal-routing of Tf in the kidney may have an indirect down-regulating
effect on the intestinal uptake of iron via the maintenance of plasma iron levels.
20
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1 3 The Recycling of Transferrin Receptors
Transferrin receptors have been studied extensively in filter-grown Madin-Darby
Canine Kidney (MDCK) cells, a well characterized model system to study vesicular
transport in polarized epithelial cells (167). In MDCK cells, endocytosis occurs
from both apical and basolateral plasma membrane (168; 169) and the basolateral
and apical endosomes appear to be interconnected (170-172). For example, Odorizzi
et al. reported the colocalization of the apically and basolaterally internalized Tf
receptors in the endosomal tubules in the cytoplasm of MDCK cells (172). The
internalized Tf receptors were transported to the basolateral membrane in a signal
dependent manner (172). However, Tf recycling in MDCK cells has been shown to
occur from both the basolateral early endosomes and the endosomes located close to
the centrioles in the cells (173; 174). Sheff et al. described two Tf recycling
pathways with different kinetics in MDCK cells (174). In their analysis, they used a
model that was defined by a series of first order rate constants for the transfer of the
ligands, such as Tf, between different compartments. The majority of Tf (65%) was
found to be recycled from the early endosomes within 10 minutes after
internalization (174). The slow phase of Tf recycling occurs from the recycling
compartment (174). A recent investigation of Tf recycling in CHO cells in which
perinuclear RE was removed microsurgically, showed the existence of distinct
recycling endosome populations in the cytoplasm (175). The endosomes appeared to
21
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
correspond to the early endosomes (EEAl-positive), and the recycling endosomes
(EEAl-negative), which recruited Rabl 1 with time (175).
1.3.1 Rab Proteins Involved in Transferrin Trafficking
Rab proteins belong to a family of monomeric GTPases with molecular masses of
20-40 kDa that are involved in different intracellular processes including regulation
of vesicular trafficking pathways (176). The small GTPases Rab4, Rab5, and Rabl 1
have been studied extensively, and Tf in the endocytotic route has been found in
structures containing Rab5, Rab4 and R abll. There are three endosomal
populations. These are characterized by structures which contain only RabS, both
Rab5 and Rab4, and both Rab4 and R abll, respectively (177). RabS, which is
localized primarily to the early endosomes, can be detected in low abundance in
Rab 11-positive recycling endosomes (178; 179). Tf colocalizes with RabS in
clathrin coated vesicles and within RabS-positive endosomes (177; 179). It was
reported that overexpression of the dominant positive mutant of RabS, RabS(Q79L)
inhibits the exit of Tf from the RabS-positive endosomes to the Rabl 1-positive
recycling endosomes (180). On the other hand, the rates of TfR-mediated
transcytosis and recycling were not affected by the overexpression of RabS(Q79L)
in Hela cells. This suggests that the GTP hydrolysis by RabS is not rate-limiting for
the endocytosis of TfR (181), and Tf receptors that are accumulated in the early
endosomes may be recycled back to the plasma membrane by the fast recycling
2 2
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
pathway. After the discovery of RabS isoforms, RabSb and RabSc, the RabS protein
was renamed RabSa (182; 183). These three isoforms share all the structural features
required for the regulation of the endocytosis. Both RabSb and RabSc increased the
rate of Tf endocytosis in vivo (182). They were also shown to stimulate the early
endosome fusion in vitro (182). However, the RabS isoforms are phosphorylated by
different kinases in vitro (184).
Rab4 is associated with early endosomes (18S) and recycling endosomes, but
not with plasma membrane (186). It appears to be less abundant on recycling
endosomes compared to early endosomes (174). In addition, it is phosphorylated by
p34cdc2 kinase (187). In MDCK cells cotransfected with human TfR and either
Rab4 or the dominant positive mutant of Rab4(Q67L), Tf receptors have been found
to redistribute from the basolateral surface to the apical surface (188). An
enhancement of the apically-directed transcytosis of Tf has also been observed in the
transfected MDCK cells and Rab4 (188). Expression of the dominant negative
mutant of Rab4(S22N) or of the chimeric NHRab4cbvn, in which the carboxyl-
terminal Cys-Gly-Cys motif of Rab4 was replaced with the transmembrane domain
of cellubrevin, did not show any effect on Tf transcytosis in MDCK cells (188; 189).
These results suggest that not only binding to GTP, but also the routing between
membrane and cytosol phases, are important for the function of Rab4. However, the
role of Rab4 in the TfR recycling is not completely understood. Chavrier et al. has
suggested that Rab4 is involved in Tf transport between early and recycling
endosomes (190).
23
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
The presence of Rabl 1-positive membranes which are enriched in actin,
annexin II and t-SNARE syntaxin 13 has been reported (179). This is consistent
with the role of actin filaments in TfR recycling (191). The polarized recycling of Tf
in MDCK cells was inhibited by latmnculin B, which depolymerizes the actin
filaments (192). In Caco-2 cells latrunculin A, a latmnculin B analog, caused 20%
missorting of pre-intemalized Tf to the apical side (193).
R abll has been detected in several different cell types, including epithelial
cells (194). The expression of Rabl 1 in the apical vesicular population in epithelial
cells was reported by Goldenring et al (194). Other studies also reported the
presence of R abll in the pericentriolar recycling endosome (180; 195; 196) and
trans-Golgi network (197; 198). Rabl 1 is also involved in the recycling of
intemalized Tf (180). Expression of a dominant negative mutant of R abll inhibited
the slow phase of Tf recychng (180). The constitutively inactive mutant of Rabl 1
(Rabl 1S25N) was shown to block the Tf recycling at both 16°C and 37°C, while the
wild type Rabl 1 and the dominant positive mutant form of Rabl 1 (Rabl 1Q70L)
inhibited the Tf recycling only at 37°C (196). Therefore, the activation of Rabl 1 by
GTP appears to be required for the exit of Tf from early endosomes towards either
the recycling endosome or the plasma membrane (196).
The Rabl 1 subfamily consists of two isoforms, Rabl la and Rabl lb, with a
homology of about 89%. Rabl lb has been shown to colocalize with intemalized Tf
in the pericentriolar recycling endosome and is essential for recycling of Tf to the
plasma membrane (199). Rab25, a small GTPase closely related to R abll,
24
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
colocalizes with R abll in MDCK cells (167). The rate of IgA transcytosis and of
apical recycling of intemalized ligands was decreased upon the overexpression of
Rab25 in MDCK cells (167). The basolateral recycling of intemalized Tf was not
affected by overexpression of Rab25, suggesting that Rab25 may play a role in
apically directed trafficking (167).
1.3.2 The R ab ll Interacting Proteins
A number of rabl 1 interacting proteins including R ipll (200), Rab Coupling Protein
(RC?) (201), Rabl 1-Family Interacting Proteins, Rabl 1-FIPl, Rabl 1-FIP2, Rabl 1-
FIP3 (202), and Rabl 1-FIP4 (203) have been identified. The interactions between
Rabl 1 and Rabl 1-FEPs primarily involve the C-terminal domains of the FBPs, which
includes the Rab binding domain (RBD) (201-204).
RCP interacts with both Rab4 and R abll. It is likely to be involved in Tf
recycling (201). Rabl 1-FIP2 and Rabl 1-FIP3 have been localized to the rabl 1-
positive compartment in MDCK and HeLa cells (202). Rabl 1-F1P2 appeared to be
involved in recycling of Tf (204). Rabl 1-FIP4 colocalizes with both Rabl 1 and Tf
in HeLa cells (203). It interacts with Rabl 1 in a GTP-dependent manner (203). The
expression of a mutant form of Rabl 1-F1P4 (rabl l-FIP4(C-ter)) did not show any
effect on Tf recycling in HeLa cells but dispersed the Rabl 1-positive compartment
toward the peripheral region of the cell (203).
25
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.3.3 RabS Effectors, EEAl and PI(3)K, Involved in Early Endosome Fusion
Early endosomal antigen 1 (EEAl), a hydrophilic 162kD protein localized to the
early endosomal membrane, plays a critical role in the fusion of early endosomes
(181; 205; 206). It has also been found as a homodimer in the cytosolic pool (206;
207). EEAl functions as a tethering protein for two RabS positive membranes (208)
which may facilitate the pairing of SNARE proteins (209) and subsequently
membrane fusion (207; 210). It contains two binding sites for RabS at the N and C
termini (208). Association of EEAl with the endosomal membrane appears not to be
dependent on its interaction with RabS (211). However, the interaction between
RabS and EEAl may regulate early endosome fusion (211). According to Stenmark
et al., the endosomal localization of EEAl requires the presence of a FYVE domain
(212), a cysteine-rich domain at the C-terminal of EEAl protein which binds two
zinc ions (20S; 212) and has a binding site for PI(3)P (213-216). The interaction
between the FYVE domain and PI(3)P is not only specific (217), but also necessary
and sufficient for the binding of EEAl to the endosomal membranes (211). PI(3)P
on the early endosomes or endocytic vesicles could be generated by the recruitment
of PI(3)kinase hVPS34 by RabS-GTP (218), which limits the recruitment of EEAl to
a specific region of the membrane (20S; 208). In addition, the interaction between
hVPS34 and RabS may be mediated by hVPS34-associated protein plSO (218).
So far, two distinct PI(3)Ks, hVPS34 and p8Sa-pl lOp, have been found to bound to
RabS (218). Class-HI PI(3)K hVPS34 is the only PI(3)K required for early
26
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
endosome fusion, and PI(3)P is the most important 3'-phosphatidylinositide in the
fusion process (218). The class-I PI(3)K, p85a-pl lOp, which produces
phosphatidylinositol-3,4-bisphosphate and phosphatidylinositol-3,4,5-trisphosphate,
may be involved in the formation of CCVs at the plasma membrane (218). The
involvement of p85a-pll0P in Tf recycling has also been demonstrated. Activation
of class-I PI(3)K by insulin receptor enhanced the rate of Tf recycling while
antibodies against the pi 10a subunit of PI(3)K inhibited Tf recycling (219; 220).
The involvement of PI(3)Ks in Tf transcytosis has also been studied by using
PI(3)K inhibitors, such as wortmannin and LY294002. Wortmannin is a
noncompetitive, irreversible inhibitor of mammalian PI(3)Ks at low concentration
(100 nM), while the inhibitory effect of LY294002 is more reversible and specific
for PI(3)Ks (221-223). The rate of the Tf recycling was reduced by Wortmannin and
LY294002 (224-227).
According to van Dam et al. Tf receptors may be recycled back to the plasma
membrane via two different independent recycling pathways that require the
activities of PI(3)K and dynamin (228). Tf receptor recycling from perinuclear
recycling endosomes was inhibited upon the overexpression of a dominant negative
dynamin-1 mutant (dynts), which accumulates clathrin-coated buds on endosomes
(229). An accumulation of endocytosed Tf in the early endosomes was observed in
the presence of LY294002 indicating that PI(3)K activity is required for the Tf exit
from early endosomes (228). No interference between LY294002 treatment and Tf
transport between either early endosome and recycling endosome or recycling
27
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
endosome and the plasma membrane was reported, suggesting that LY294002 may
inhibit the Tf recycling from the early endosomes to the plasma membrane (228).
The combination of LY294002 and BFA, which appeared to inhibit the assembly of
clathrin coats on recycling endosomes, had a synergistic inhibitory effect on Tf
recycling (228).
1.4 Utilization of TfR for drug delivery
TfR targeted therapy has emerged as an interesting drug-delivery tool with dual
functionality. For example, targeting of TfR can lead to delivery of therapeutic
agents into tissues of choice or across epithelial barriers of choice. The seemingly
contradictory effects can be achieved by focusing the targeting strategies toward
different aspects of TfR-related biology. This section focuses on recent advances in
TfR-facilitated drug delivery.
1.4.1 TfR-based targeting for anti-cancer therapeutics
One of the most well explored avenues for TfR-based drug targeting strategies is the
use of anti-cancer based therapeutics conjugated to Tf (or TfR recognizing antibody)
to preferentially direct the drug to TfR-rich cancer cells. High levels of TfR
expression have been demonstrated in many tumors (108),(230-234) and importantly,
studies have also shown that the TfR is expressed more abundantly in malignant
28
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
tissues than their normal counterparts (108; 115; 233; 235). TfR is also more
abundantly expressed in rapidly dividing cells than quiescent cells (236; 237)
because of its pivotal role in iron uptake and the absolute requirement for iron in
rapid cell proliferation. Therefore, the TfR expressed on cancer cells has been seen as
a suitable target for the delivery of therapeutics by receptor-mediated endocytosis.
Many anti-cancer agents have been considered for conjugation to Tf by
varying methods, including direct chemical linkage (238; 239), liposomal packaging
of toxin and linkage to Tf (240; 241), conjugation of DNA/polylysine complexes to
Tf (242-245), and conjugation of liposome/DNA complexes to Tf (246-252).
A great variety of cytotoxic agents have been conjugated to Tf and
investigated as potential anti-cancer therapeutics. Some of the more notable
examples have been methotrexate (238; 240), doxorubicin (238; 239), cisplatin
(253), ricin A (254-257), daunorubicin (241; 258), and toxin CRM107 (259; 260).
Conjugation of these toxins to Tf has the dual benefits of reducing toxicity in
undesired tissues and increasing the targeting efficiency to the cancerous cells. Of
special note is that conjugation to Tf significantly enhances the effectiveness of these
agents in many multi-drug resistant cell lines. For example, Tf-doxorubicin was 5 to
10 times more effective than doxorubicin-control in killing doxorubicin-resistant cell
lines, whereas in doxorubicin-sensitive cell lines, the conjugate was only 4 to 5 times
more effective than control (261). The Tf-based enhancement effect becomes even
more dramatic as one considers the very highly resistant KB cell line. In this case,
the Tf-doxorubicin conjugate exhibited an IC50 value as low as 0.025 pM, while the
29
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
un-conjugated doxorubicin demonstrated no cytotoxic effects with concentrations as
high as 1 |iM (262). The exact mechanism by which Tf conjugation allows
doxorubicin to effectively bypass the drug-resistance machinery of the resistant cell
lines is not fully understood; however, it is conceivable that internalization of the Tf-
doxorubicin conjugate allows for sequestration into the endocytic pathway, away
from drug efflux proteins (such as PGP and/or MDR), which normally reside at the
plasma membrane.
A variation on the theme of using toxins conjugated to Tf has been
investigated to increase efficiency of uptake at the cellular surface. This approach
relies on the formulation of the toxin in liposomal delivery vehicles to further
enhance the efficiency of cellular uptake. For example, methotrexate that has been
formulated in a liposome complex and conjugated to Tf demonstrated a higher
efficiency of cellular uptake and cytotoxicity compared to free methotrexate and
methotrexate-liposome control (240). In a similar fashion, when daunorubicin-
containing liposomes are conjugated to 0X26 ant-rat TfR antibody, they show an
increased delivery and uptake in the CNS relative to the non-OX26 containing
liposomes (241).
An alternate approach toward treating cancerous tissues relies upon gene
therapy to correct errors in genes, such as the tumor repressor gene p53, rather than
killing the cell. Tf-conjugation has been investigated as means to overcome some of
the limitations of cationic liposome-based p53 gene delivery vehicles, such as poor
stability in serum, binding to serum proteins, and lack of specific targeting (263-
30
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
267). Tissue culture experiments have revealed that the Tf conjugate significantly
enhanced the transfection efficiency of the p53 gene in cancerous cell lines
compared to the control DNA-liposome (265). This enhancement was seen even in
the presence of high concentrations of serum. In addition, experiments with nude
mice containing subcutaneous DU-145 tumors revealed that administration of Tf-
liposome conjugate was sufficient for high levels of p53 gene expression in the
tumor. This contrasts with the non-Tf bearing p53-liposome which did not
demonstrate any p53 gene expression in the tumor (267). This strategy has even
developed to the point where beneficial effects are seen in human patients. For
example, when a cationic liposome (carrying p53 DNA) was investigated as a Tf
conjugate, it demonstrated high efficiency in p53 gene delivery to human head and
neck, prostate cancers, as well as long-term therapeutic benefits. For example, when
Tf-liposome therapy was combined with conventional radiation therapy, complete
tumor regression was observed in human prostate cancer and the treatment group
showed no signs of relapse up to six months later (266). Using similar plasma
membrane permeability-enhancing logic as the DNA/liposome complexes, reports
have also focused on DNA/polycation complexes conjugated to Tf to increase
uptake. Comparable benefits in enhanced uptake have been observed for the
polylysine and polyethylenimine containing complexes (242; 245; 268-273). Further
developments in Tf-based targeting of anti-cancer drugs will most likely play
important roles in anti-cancer therapy in the future.
31
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.4.2 TfR-mediated transepithelial protein-drug delivery
Modem biotechnology has produced many potent and beneficial drags, yet most of
the products suffer from limited choices regarding potential routes of administration.
The large sizes and charges of protein drags prevents their passage across epithelial
barriers. TfR has emerged as a potential mediator to enable the transport of these
large molecules across epithelial barriers.
1.4.2.1 anti-TfR antibody drug conjugates for transepithelial drug delivery
The capillaries of the brain have been well known to have relatively high levels of
TfR, and since the blood brain barrier (BBB) effectively excludes many therapeutic
drags, including almost all peptide and protein-based therapeutics, targeting of the
BBB’s TfR for drag delivery has been seen as an attractive strategy. However,
utilizing a Tf-based drag conjugation approach is seen as undesirable for trans-BBB
delivery because under physiologic conditions the TfR of the BBB are postulated be
nearly saturated with Tf, due to the relatively high amounts of Tf in serum (274). A
viable altemative to target BBB TfR has been shown to be antibodies that
specifically recognize TfR, with or without the presence of bound Tf (275-278). In
particular, 0X26, a monoclonal antibody for rat TfR that has been the subject of
most of the research in this area, demonstrated an ability to preferentially target the
32
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
TfR of the BBB (111). OX-26 is postulated to not interfere with the normal
functioning of iron uptake via TfR, since the antibody binds an extra-cellular region
that is distinct from the binding pocket for Tf (111).
Upon binding to TfR at the BBB, 0X26 exhibits superior transport abilities
into the CNS. Studies performed with isolated bovine brain capillaries demonstrated
that nearly 50% of bound radiolabeled 0X26 was taken up via endocytosis during a
two hour incubation period (279). In addition, carotid artery perfusion studies
indicate that more than 65% of the administered 0X26 passed through the BBB and
into the extracellular space. This value was significantly higher than for the control
proteins, bovine serum albumin and IgG, thus implying that transcytosis via TfR
significantly enhanced the transport of 0X26 across the BBB. Intravenous bolus
administration of 0X26 also resulted in a steady accumulation of radiolabeled 0X26
in the brain during a five-hour time period, while radiolabeled 0X26 was rapidly
cleared from serum after injection. This suggests that 0X26 rapidly binds TfR at the
BBB and is subsequently endocytosed across the BBB in a more gradual fashion
(279).
The next logical course of action was to determine if 0X26 could enable the
efficient CNS uptake of therapeutics that would normally exhibit poor CNS-
bioavailability. The strategy most commonly reported in the literature involved the
creation of OX26-therapeutic conjugates based on strepavidin/biotin linker
technology. The avidin/biotin linkage between 0X26 and the therapeutic entity has
been created using chemical based and genetic-cloning based strategies. Several
33
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
promising 0X26 conjugates have been created, using a vasoactive intestinal peptide
analog (280), brain-derived neurotrophic factor (BDNF) (281-283), and basic
fibroblast growth factor (bFGF) as the therapeutic cargo. (284; 285). Depending
upon the pharmacokinetic properties desired, the conjugates have been prepared with
avidin-modified 0X26 biotin conjugated to the drug directly or via a PEB 2000
spacer. When BDNF-PEG2000-SA-OX26 was administered to rats for one week
after an induced episode of transient forebrain eschemia, a neuroprotective effect
was observed. Specifically, the neuronal density in the hippocampus of the BDNF-
PEG2000-SA-OX26 treatment group was maintained, while the group receiving pure
BDNF exhibited a 68% decrease compared to control (286). In addition, BDNF-
0X26 conjugate demonstrated effectiveness in ameliorating the effects of acute
stroke. For example, when adult rats were subjected to 24 hours of middle cerebral
artery occlusion, the infarct volume was found to decrease in a BDNF-OX26 dose
dependent fashion. Reduction in stroke volume was significant even if
administration of BDNF-OX26 was delayed for one to two hours after the occlusion
event (281; 282; 287; 288).
bFGF is an attractive drug candidate for 0X26 conjugation because it retains
receptor-binding affinity and has increased brain uptake following conjugation to
OX26-SA. Studies also have demonstrated that bFGF-OX26 conjugate protects
cortical cell cultures against hypoxia/reoxygenation insult in a dose-dependent
manner in vitro (285). In addition, in vivo studies showed that a single intravenous
injection of bFGF-OX26, equivalent to a dose of 25 pg/kg bFGF, produces an 80%
34
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
reduction in infarct volume in the brains of rats subjected to permanent occlusion of
the middle cerebral artery. This result paralleled with a significant improvement of
neurologic deficit. The neuroprotective action was also time-dependent, with a 67%
reduction in stroke volume if bFGF-OX26 was administered 60 min after arterial
occlusion, whereas no significant reduction was observed if treatment was delayed
by 2 h (284; 285).
In addition to the delivery of large therapeutic molecules across the BBS,
TfR based transcytosis has also been explored for targeted gene delivery to the CNS.
For example, high levels of reporter gene expression have been observed when
plasmids encoding for luciferase or p-galactosidase are enclosed in liposomes that
are conjugated to 0X26 via a PEG2000-thio-ether linkage were administered
intravenously to rats (289-291).
1.4.2.2 Enhancement of TfR-mediated drug delivery
In addition to the use of Tf-drug conjugates to target therapeutics to select tissues
from the blood stream, Tf based conjugates have also been considered for the
systemic non-invasive delivery of therapeutics. Several absorptive epithelia have
been shown to express TfR, such as the small intestine (112) and type II
pneumocytes (114), providing a potential target to enable transepithelial delivery of
large bio-active molecules that would normally have negligible bio-availability when
administered orally. One of the problems associated with this approach is the fact
35
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
that most of the surface TfR in differentiated epithelial cells lies at the basolateral, or
serosal, membrane (292). This would tend to preclude the ability to achieve apical-
to-basolateral transcytosis of Tf-drug conjugates. While there is indication that
transepithelial transport may still be feasible (especially in areas where large
numbers of semi-differentiated cells may occur, such as the crypt of Lieberkuhnin of
the small intestine), enhancers of Tf-conjugate transepithelial transport have been
examined as a means to overcome the problem of basolateral-localized TfR polarity.
1.4.2.2.1 Lysosomotrophic Amines
Lysosomotrophic amines (such as ammonium chloride and chloroquine) have been
considered as a means to enhance Tf-drug conjugate transepithelial transport. Most
of these agents increase intraendosomal and intralysosomal pH, with a concomitantly
enhanced accumulation of radiolabeled Tf inside the cell. However, these agents also
seem to cause a non-specific enhancement of ^^^Tf transcytosis. For example when
MDCK monolayers were treated with NH4CI (20 mM), a 7-fold increase in
basolateral-to-apical transcytosis was observed. The increase in transcytosis was
thought to be derived from the overloading of labeled ^^Tf inside the cell (293). The
serious drawback to these agents is the nonspecific alteration of membrane
permeability and integrity.
36
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.4.2.2.2 Lipophilic carboxylic ionophores
Carboxylic ionophores, such as monensin and nigericin, exhibit effects similar to
lysosomotrophic amines in altering the pH of endosomal and lysosomal
compartments (294). Monensin, is a Na'^-ionophore that can elicit a variety of
effects in mammalian cells, including alterations of vacuolization and vesicle sorting
in the Golgi and trans-Golgi networks. Treatment of MDCK monolayers with 6 pM
monensin resulted in a 24-fold increase in TfR-mediated transcytosis. The large
increase in TfR-mediated transcytosis appeared to originate from a different mode of
action than lysosomotrophic amines. This was presumed to result from the effects of
monensin on trans-Golgi sorting, since TfR is routed to the TGN as a part of the
normal course of its intracellular sorting (295; 296). The development of monensin
as a useful therapeutic agent has been hampered by the highly lipophilic nature of the
drug, which is reflected in its poor stability and potentially short serum half-life.
1.4.2.2.3 Brefeldin-A
Another transcytosis enhancing reagent that has been shown to have an effect in
modulating the trans-Golgi network is the fungal metabolite, brefeldin-A (BFA).
BFA has been extensively used in the study of intracellular trafficking events (297).
It has dramatic effects on the structure and function of the Golgi complex and the
endosomal sorting pathways (298-300). For example, BFA causes the rapid
37
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
rearrangement of the Golgi, through microtubule dependent processes, causing
Golgi-specific markers to redistribute in a retrograde fashion back to the
endoplasmic reticulum (301; 302). In addition, the trans-Golgi network tubulates in
the presence of BFA, forming a fused trans-Golgi/endosomal network that appears to
exclude vesicles that are lysosomal in nature (298; 303). While the lysosomes do not
appear to fuse with the trans-Golgi/endosomal tubulates, they do appear to form an
extensive microtubule dependent network in the presence of BFA (298). In addition
to these effects, BFA also blocks the anterograde transport of sorted proteins from
the endoplasmic reticulum (301; 302). Unlike the reagents discussed above, BFA
does not cause a change in intra-lysosomal or intra-endosomal pH (298).
In polarized epithelial cells, BFA treatment significantly up-regulates the
basolateral-to-apical transcytosis of internalized TfR and bulk plasma membrane
(304). In spite of these observations, the Golgi of some cell lines seem to be
resistant to the effects of BFA. For example, the Golgi of MDCK cells largely
resists the effects of BFA (305). Nonetheless, BFA causes significant alteration of
trans-Golgi and endosomal function in MDCK cells (305). The fact that BFA causes
significant enhancement of TfR-specific transcytosis in this cell line would indicate
that this enhancement depends upon sorting events at the TGN/endosome level.
BFA significantly alters the intracellular sorting of internalized proteins in
polarized epithelial cells. For example, BFA treatment results in the missoiting of
basolateral LDL receptors (306), reduces the efficiency of polymeric immunoglobin
receptors to the cell surface (307). In addition, the effects of BFA on protein sorting
38
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
appear to be much more sensitive at the apical surface, as the concentration of BFA
required to see effects at the apical surface is much lower than for the basolateral
surface (308; 309). This is significant when considering BFA as an enhancer of TfR-
mediated drug delivery. The relative preferential alteration of events at the apical
surface would imply the ability to achieve net enhancement of TfR-mediated
transcytosis in the apical-to-basolateral direction. This effect has been demonstrated
in two highly polarized cell systems, MDCK cells (292) and primary cultured rat
type n pneumocytes (310). In both of these systems, BFA treatment results in the
specific enhancement of apical-to-basolateral transport of conjugates of Tf-insulin
and Tf-GCSF (filgrastim) (292; 310). The rates of apical-to-basolateral transport of
the conjugates are significantly higher in the presence of BFA than for comparable
molar concentrations of the native protein. The enhancement of transport appears to
result from the transient missoiting of basolateral TfR to the apical surfaces of the
cells, from whence they are rapidly intemalized and redirected back to the
basolateral membrane, releasing the Tf-protein-drug conjugate into the basolateral
fluid. This is evidenced by results which demonstrate that BFA-treatment does not
increase the static level of TfR at the apical surface, as measured by *^^I-Tf binding
at 4° C, but rather increases the rate of TfR internalization, as measured by ^^Fe-Tf
uptake. While BFA does affect the trans-epithelial electrical resistance (TEER) of
the monolayers, the effect is transient and TEER retums to normal levels once the
BFA is removed (292; 310).
39
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.4.3 Modification of Rab-dependent sorting pathways for enhancement of
TfR-mediated gastrointestinal transepithelial transport
While investigations into BFA-induced enhancement of TfR-mediated
transport have demonstrated a significant elevation in Tf-protein-drug conjugate
transport in polarized epithelia, the fact that BFA is a fungal metabolite makes it
undesirable in some respects. For example, BFA affects many properties of
intracellular organelles, even those that might be unrelated to TfR-based sorting or
transcytosis events. The non-specific side effects of BFA prompted efforts to
discover agents that would have the potential to be more specific in their mode of
enhancement of TfR-mediated transcytosis. As discussed in section 2, several small
GTPases of the Rab family have been shown to be involved in the endocytosis and
transcytosis of TfR. Therefore one can pose the hypothesis that chemical entities
that inhibit or alter the actions of key Rabs, or their downstream effectors, could
possibly control the sorting and recycling events of TfR to elicit a specific
enhancement of TfR-transcytosis.
Recent reports have focused on the ability of AG-10 (aka tyrphostin- 8 [T-8 ],
or 4-hydroxybenzylidene-malononitrile), a known GTPase inhibitor, to increase the
transport of an insulin-Tf conjugate across Caco-2 monolayers (in the apical-to-
basolateral direction) and enhance the oral absorption of the conjugate in
streptozotocin-induced diabetic rats across the GI epithelium (311; 312). In Caco-2
monolayers, treatment with AG-10 induced a 20-fold enhancement of TfR-mediated
40
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
transport of insulin (via the insulin-Tf conjugate) compared to control, while similar
treatment with BFA only resulted in a 3-fold enhancement of insulin transport (312).
This enhancement of transport was determined to be dependent upon TfR-mediated
processes, since an insulin-albumin conjugate demonstrated no transport across the
monolayers, and treatment with AG-10 had no effect on its transport properties
(312). The results from the tissue-culture experiments have been further validated by
in vivo uptake studies. In diabetic rats, the orally administered insulin-Tf conjugate
demonstrated an extended duration of action, with hypoglycemic effects observed 11
hours after administration (113). In addition, AG-10 exhibited the ability to enhance
the hypoglycemic effect of orally administered insulin-Tf conjugate, relative to BFA-
treated and enhancer-free insulin-Tf treatment groups. The effects of AG-10 were
also dose dependent, indicating the observed results resulted from of the effects of
AG-10 (312).
In an effort to further increase the specificity of enhancement of TfR-
mediated transcytosis, downstream effectors of Rab proteins have been considered as
potential targets for inhibition. One of the downstream effectors of Rab proteins are
the phosphatidylinositol 3-kinases (PDK). As noted earlier, PDK’s are required for
cellular membrane trafficking and they have been implicated in the recycling of TfR.
For example, the PDK inhibitors, LY294002 and wortmannin, have been shown to
reduce the rate of Tf recycling (224-227; 313; 314), whereas activation of PDK
activity is known to accelerate the rate of TfR recycling (315; 316). One can
hypothesize that inhibition of recycling of basolaterally derived TfR may result in an
41
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
increase of TfR txaffic to the apical membrane, thus increasing the potential for
enhancement of TfR-mediated transcytosis. Recent results have suggested that
LY294002 is capable of enhancing TfR-mediated transcytosis in a manner that is
similar to AG-10 and different from the mode of action of BFA. For example,
LY294002 specifically enhanced TfR-mediated transcytosis of ^^^I-Tf and ^^^I-Tf-
GCSF conjugate in Caco-2 monolayers, yet demonstrated less of an effect in MDCK
monolayers. This contrasts with BFA, which has much greater enhancement ability
in MDCK monolayers compared to Caco-2 (292; 312). Caco-2 and MDCK are
known to have distinct TfR-sorting pathways, i.e. predominately direct TfR sorting
in MDCK cells, and both indirect and direct sorting in Caco-2 (312). This suggests
that AG-10 and LY294002 affect one type of sorting mechanism, while BFA affects
another. This hypothesis has been supported by a recent report, which demonstrated
that endocytosed TfR are recycled via distinct dynamin and PDK dependent
pathways, where BFA and LY294002 exhibited distinct yet synergistic inhibitory
effects (228).
1.4.4 Enhancement of TfR-mediated transport in pulmonary epithelium.
The large surface area of the lower respiratory tract presents a challenge to
pulmonary host defense. In contrast to upper airways, the epithelium of alveoli is
devoid of the protective mucus barrier and the ciliated cells that enable the constant
clearance of foreign material in the upper airways. While alveolar macrophages may
42
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
provide a formidable first line of defense against pathogenic organisms, of particular
concern is the presence of excessive levels of free iron in the surface fluid lining the
alveolar epithelium in the distal lung (317). Increased levels of free iron in the distal
lung (as a consequence of pathological conditions such as smoking or chronic
inhalation of metallic dusts) have been associated with tissue injury and fibrosis,
primarily due to iron's ability to catalyze the formation of highly reactive hydroxyl
radicals (317-319). Excessive levels of free iron in the lower lung have also been
associated with facilitating the growth of intracellular mycobacteria (318; 320-322).
The task of sequestering free iron lies with the iron-binding proteins, of
which Tf is the primary constituent in the surface fluid lining the airspaces of the
lower lung (323). Tf is an 80 kDa glycoprotein that is responsible for transporting
iron, via its two ferric binding sites, throughout the vasculature (324). In addition to
this primary role, Tf has been implicated as the major source of antioxidant capacity
in the lung (325). Indeed, the levels of Tf, as a percentage of total protein, detected
in bronchoalveolar lavage fluid have been found to be very high (4.0-5.6%) as
compared to values for plasma (3.0%) (326; 327). The presence of such high levels
of apo-Tf in surface fluid lining the alveolar epithelium suggests that there may be a
specialized transport mechanism for Tf from the systemic circulation to the alveolar
fluid. The very tight epithelial barrier of the alveoli excludes a passive paracellular
transport route for the large Tf molecule (328), consistent with a hypothesis that the
transport of Tf into, and possibly out of, alveolar lining fluid might be regulated by
an active transcellular transport process. Endocytosis of Tf by the TfR and
43
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
subsequent transcytosis into alveolar lining fluid would be a likely mechanism for
this process. However, the extent and nature of TfR distribution among the cells of
the rat alveolar epithelium remain unknown.
Primary cultures of rat type II alveolar epithelial cells (AEC) have been
shown to exhibit many of the properties of alveolar epithelium in vivo, including
apparent transdifferentiation from the type II cell-like to the type I cell-like
phenotype within 3-4 days in culture (329-332). Type I pneumocytes comprise
nearly 97% of the surface area of the pulmonary alveoli, yet they are slightly
outnumbered by the more numerous type II cells. The thin expansive type I cells are
thought to primarily serve to enable efficient gas exchange, while the cuboidal type
n cells exhibit essential secretory functions, such as the secretion of pulmonary
surfactant and surfactant proteins.
As a part of this thesis, we investigated the synthesis and distribution of TfR
in polarized alveolar epithelial cell monolayers. Primary cultures of rat alveolar type
n cells were used as a model system to study TfR production in type II cell-like and
type I cell-like monolayers. Under normal culture conditions, the type II
pneumocytes progress toward the type I cell-like phenotype as the cells develop tight
junctions and the monolayers develop high transepithelial electrical resistance
(TEER) (329). This transition to the type I cell-like phenotype early in the culture
period had retarded development of a model system for studying the transport
properties of monolayers with the type II cell-like phenotype. However,
keratinocyte growth factor (KGF) has recently been shown to cause retention of the
44
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
type n cell-like phenotype in culture and prevent progression to the type I cell-like
phenotype while retaining high TEER levels (333-336). KGF exposure of AEC
monolayers was used in our study as a means to investigate the degree of TfR
synthesis in cells exhibiting the type II cell-like phenotype.
The pulmonary epithelium has become an attractive target for systemic
delivery of protein based therapeutics. The large surface area (~ 100 m^) and
extensive vascularization in the lung provide an ideal site for drug absorption.
Several protein drugs have been reported to have relatively high bioavailabilities
when delivered via the pulmonary route. However, conclusive knowledge regarding
the mechanisms of transport through the pulmonary epithelial barrier remains
elusive. In light of this, we have sought to develop a pulmonary delivery strategy
that is targeted to a known transcytosis pathway, namely, transcytosis of the TfR, in
order to obtain more predictable delivery qualities suitable for protein drugs of
varying characteristics.
TfR-mediated drug delivery has advantages over other protein drug delivery
strategies. For example, in contrast to penetration enhancers, TfR transcytosis
neither alters the cellular plasma membrane nor dismpts the cellular tight junctions.
As TfR endocytosis is an endogenous transport process, it is probable that a protein
drug delivery scheme targeting TfR would have fewer side effects compared to other
methods. In addition, Tf is an advantageous protein carrier molecule because it is
inherently resistant to enzymatic degradation processes (337-339). Primary cultures
of rat type II alveolar epithelial cells (AEC) have been shown to exhibit many of the
45
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
properties of alveolar epithelium in vivo, including apparent transdifferentiation from
the type II cell-like to the type I cell-like phenotype within 3-4 days in culture (329-
332). Type I pneumocytes comprise nearly 97% of the surface area of pulmonary
alveoli, yet they are slightly outnumbered by type II cells. In addition to active N a"^
absorption afforded by both types of pneumocytes (340-342), the thin expansive type
I cells are thought to primarily serve to enable efficient gas exchange, while the
cuboidal type II cells exhibit essential secretory functions, such as secretion of
pulmonary surfactant and surfactant proteins.
Primary cultured type II pneumocytes usually acquire the type I cell-like
phenotype and morphology as the cells develop tight junctions leading to monolayers
with high transepithelial electrical resistance (TEER) (329). This transition to the
type I cell-like phenotype had retarded development of a model system for studying
transport properties of monolayers with the type II cell (-like) phenotype. In this
regard, keratinocyte growth factor (KGF) has recently been shown to cause retention
of the type H cell-like phenotype in culture, while retaining high TEER levels (333-
336). Thus, KGF-treated AEC monolayers were used in this study as a means to
investigate the degree of TfR transcytosis in cells exhibiting the type II cell-like
phenotype.
Our first report (114) demonstrated that TfR expression is primarily
limited to AEC monolayers of the type II cell-like phenotype, and that TfR
expression is predominantly localized to the basolateral membrane. KGF treatment
(10 ng/mL) of AEC monolayers was found to restore TfR expression and TfR-
46
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
mediated Tf endocytosis. Later studies utilized these previous findings to develop a
TfR-mediated protein drug delivery strategy that targets TfR in type II cell-like AEC
monolayers. (343)
TfR has developed as a potential ligand to enable drug targeting and delivery
of therapeutic agents that would normally suffer from poor pharmacokinetic
characteristics. TfR-directed targeting has enabled the efficient delivery of
therapeutic agents to sites of interest, including the central nervous system and
malignant tissues. In addition, by utilizing knowledge of the intracellular sorting and
recycling pathways of TfR, including Rab and PI(3)K mediated processes, one can
maximize the transepithelial delivery of peptide-based therapeutics. Depending
upon the desired result, apparently paradoxical effects can be achieved. For
example, TfR-based strategies can achieve accumulation of the carried-drug within
targeted tissues or delivery of the therapeutic entity across tissues of interest. Further
understanding of the intra-cellular events that govern the destiny of intemalized TfR
will result in ever increasing interest in TfR as a pharmaceutically relevant molecule.
47
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2. MATERIALS AND METHODS
2.1 Caco-2 Cell Culture
Caco-2 cells (American Type Culture Collection, Rockville, MD; passage
number 20) were grown on 0.4 pm pore-size polycarbonate Trans well (Costar,
Cambridge, MA) filters to confluence in Dulbecco’s modified eagle media
supplemented with 10% fetal bovine serum (FBS), glutamine (2 mm), non-essential
amino acids (0.1 mM), penicillin (50 U/mL) and streptomycin (50 pg/mL). All cell
culture reagents were obtained from Invitrogen/Life Technologies (Carlsbad, CA).
The Caco-2 cells were typically seeded at 4.2 x 1 0 "^ cells/cm^. The monolayers were
also allowed to differentiate for seven days after reaching confluence, which was
typically 7-8 days post-seeding. The cells were grown in a 5% CO2 in air tissue-
culture incubator at 37° C. Media containing 10% FBS was changed every other
day. The trans-epithelial electrical resistance was measured with an epithelial
voltohmmeter (EVOM, World Precision Instruments, West Haven, CT). The
monolayers typically established maximum resistances of 500 Q®cm^.
2.2 MDCK Cell Culture
MDCK cells were cultured in polycarbonate Trans well filters (0.4 pm pore
size) in a 5% CO2 in air tissue-culture incubator at 37° C. The cells were seeded at a
density of 2.2 x 10“ * cells/cm^ in minimal essential medium (MEM) containing 10%
48
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
FBS, glutamine (2 mm), penicillin (50 U/mL) and streptomycin (50 p,g/mL). In
order for the cells to properly develop tight junctions, a feeding protocol was used
that sequentially reduced the percentage of FBS within the culture media. For
example, after the initial seeding, the cells were allowed to grow for three days in
10% FBS containing MEM and then the media was changed to MEM with 2.5%
FBS for two days with daily feeding, and then changed to 1% FBS containing MEM
prior to using the monolayers in transport experiments. The monolayers were used
at day 7 to 8 post seeding, on average. The TEER was typically 2.5 to 3.0 kO®cm^
when the monolayers were used in transepithelial transport studies or TfR
distribution studies.
2.3 Primary Culture of Rat Alveolar Epithelial Cell Monolayers
Lungs of specific pathogen-free Sprague-Dawley male rats were perfused via the
pulmonary artery and lavaged with Ca^'^/Mg^'^ -free Ringer’s solution. The lungs
were then instilled with porcine pancreatic elastase (2.5 U/mL, Worthington
Biochemical, Lakewood NJ) for 20 minutes at 37°C. Minced lung tissue blocks were
sequentially filtered through 100, 40, and 10 |xm Nitex membranes. Alveolar
macrophages were removed from the crude cell mixture by IgG panning (344).
Partially purified type U pneumocytes were plated onto tissue culture-treated 12 mm
polycarbonate filter (Transwells, 0.4 fxm pore size. Costar Coming, Cambridge, MA)
at a density of 1.5 million cells/cm^ in culture medium composed of a 1; 1 mixture of
49
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
DMEM and Ham’s F-12 (Sigma Chemical, St. Louis, MO) supplemented with 10%
newbom bovine serum, 1.25 mg/mL bovine serum albumin, 100 U/mL penicillin,
and 100 ng/mL streptomycin. Monolayers were fed on days three and five (or six)
with fresh culture medium. When KGF (hKGF, Calbiochem, San Diego, CA) was
added to the media bathing the AEC monolayers, it was included from day zero
onward at a concentration of 10 ng/mL and replenished with each feeding. The
TEER was measured with an epithelial voltage-ohm meter (EVOM, World Precision
Instruments, Sarasota, FL). Rat AEC monolayers typically achieved TEER > 2,500
Ocm^ and PD values of 10.2 ± 0.5mV. Animal experiments were compliant with the
Principles of Laboratory Animal Care' (NIH Publication #85-23) and approved by
the lACUC at USC.
2.4 Measurement of Cell Surface Transferrin Receptor on Caco-2 Monolayers
Human iron-loaded Tf (Sigma) was radiolabeled with (ICN, Irvine, CA) using
chloramine-T catalyzed modification (345), followed by purification by Sephadex G-
50 column chromatography and subsequent dialysis in phosphate buffered saline
(PBS, pH 7.8). Caco-2 monolayers were washed once with serum-free culture media
(DMEM) supplemented with 0.1% BSA and incubated at 37°C in the same media for
one hour to ensure removal of endogenous Tf. Monolayers were subsequently
treated with BFA (1.0 pg/mL), AG-10 (500 pM), or LY294002 (100 pM) and
incubated at 37° C for two hours to allow for uptake of the drugs. Transcytosis
50
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
enhancer-containing media were then removed and replaced with fresh media
containing 1.5 |ig/mL in either the apical or basolateral compartment and
incubated at 4°C for two hours. Non-specific binding was measured in parallel by the
simultaneous addition of 100-fold excess unlabeled Tf in the respective bathing
fluid. TfR-specific ^^^I-Tf binding was determined as the difference between total
and nonspecifically bound ^^^I-Tf. After incubation, monolayers were washed three
times with ice-cold PBS, pH 7.4. ^^^I-Tf bound to the monolayers was measured
with a Packard Gamma Counter (Packard Instruments, Meriden, CT).
2.5 Measurement of Cell Surface Transferrin Receptor in Rat Alveolar
Epithelial Cell Monolayers
Human iron-loaded Tf (Sigma) was radiolabeled and purified as mentioned above.
AEC monolayers were washed once with serum-free culture media (MDSF)
supplemented with 0.1% BSA and incubated at 37°C in the same media for one hour
to ensure removal of endogenous Tf. Monolayers were subsequently dosed with
media containing 1.5 (ig/mL '^^I-Tf and incubated at 4°C for two hours to determine
total surface receptor binding. Non-specific binding was measured as above. TfR-
specific '■^^I-Tf binding was determined as the difference between total and
nonspecifically bound ^^^I-Tf. In experiments where apical or basolateral binding
was measured, dosing of *^^I-Tf was restricted to that respective bathing fluid, as
mentioned previously. In experiments that determined total surface binding, both
51
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
apical and basolateral compartments were dosed with After incubation,
monolayers were washed as above and radioactivity associated with the monolayers
was measured with a Gamma Counter.
2.6 Measurement of Total Cellular Transferrin Receptor in Rat Alveolar
Epithelial Cell Monolayers
Total cellular TfR was determined using the method described by Lamb et al. (346).
Briefly, solubilized total cellular TfR was prepared from AEC monolayers by
detaching the cells using 0.1% Triton X-100 in 10 mM Tris-HCl, 150 mM NaCl, pH
8.0 and homogenizing the mixture by twenty passages through a Balch-press
homogenizer. Solubilized TfR was incubated for two hours on ice with 3.2 pg/mL
i25i_Tf, with or without 100-fold molar excess unlabeled Tf. ^^^I-Tf /TfR complexes
were precipitated by addition of equal volumes of 2.0 mg/mL IgG and 45%
(NH4)2S0 4 . Radioactivity in the precipitate was measured using the Packard Gamma
Counter. In addition, specific TfR binding was determined as in the surface receptor
studies. Total protein content was measured with a Pierce BCA protein assay kit.
52
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2.7 TfR-Dependent Uptake of ^® Fe in Caco-2 and Rat Alveolar Epithelial Cell
Monolayers
Human Apo-Tf (Sigma) was loaded with (Perkin Elmer, Wellesley, MA) as
previously described (347). AEC monolayers were washed as in the binding
studies described above. Monolayers were treated with BFA (l|ig/mL) in the apical
compartment and subsequently dosed with 2 jig/mL ^^Fe-Tf on either the apical or
basolateral surface. Non-TfR-mediated ^^Fe uptake was determined by parallel
experiments that included 1 0 0 -fold molar excess of unlabeled holo-transferrin in the
apical fluid. Caco-2 monolayers were incubated for six hours with ^^Fe-Tf and then
washed three times with ice-cold PBS. Cell monolayers were excised and associated
^® Fe was counted with a Packard Gamma Counter to determine uptake.
2.8 Inununoflouresence Studies in Rat Alveolar Epithelial Cell Monolayers
AEC monolayers were fixed with 3.7% formaldehyde at 18 hours post seeding
(dayl) or day 7 of culture and quenched with 50 mM NH4CI. The monolayers were
stained with mouse monoclonal antibody OX-26 (courtesy of W. Pardridge, UCLA),
which is specific for rat TfR, in conjunction with the secondary antibody, FTTC-
labeled goat anti-mouse immunoglobulin (Sigma). The monolayers were
subsequently excised and mounted, apical side up, onto glass slides with Prolong
Antifade Mounting medium (Molecular Probes, Eugene, OR). Samples were imaged
53
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
with a Nikon PCM Quantitative Measuring High-Performance Confocal System
equipped with a filter for FTTC attached to a Nikon TE300 Quantum upright
microscope. Confocal images were compiled using Adobe Photoshop 5.0.
2.9 Synthesis of Insulin-Transferrin Conjugate
Recomhinant human insulin (Sigma) was covalently linked to iron-loaded Tf using
the disulfide bifunctional linker, N-succinimidyl 3 -(2-pyridyldithio) propionate
(SPDP) according to the reaction scheme shown in Figure 4. Insulin has three amine
groups that are susceptible to modification by SPDP. In order to obtain a defined
product, the reaction scheme utilizes the difference in pKa between the two a-amino
groups of the N-terminal residues of the A and B chain of insulin and the 8 -amino
group of lysine B29 (see Figure 4). For example, the two a-amino groups were
blocked by adding 5 mg of dimethylmaleic anhydride (DMMA) three times over the
course of 30 minutes (once every 10 minutes) to a mixture of 10 mg of insulin in one
mL of 0.1 M NaHCOs. The reaction was held at pH 6 .8-7.0 to preferentially target
the a-amino groups of insulin. Subsequently, the pH was adjusted to 9.0 and SPDP
(2.5 mg in dimethyl formamide) was added and allowed to react with the e-amino
group of insulin for 1 h at 37° C. Excess SPDP was then removed from the reaction
mixture by overnight dialysis against PBS (pH. 8.0) at 4° C. The ratio of insulin to
PDP was spectrophotometrically determined to be 1 : 1.
54
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(H V M i) . SPHP
* ¥ « 3y V A ' ^ OVf^BNGIyAA^ IM V H rovA s » ^
IiKiiliii Irs-HIP
1 3 - ^ - 2 " ^ ^.MTOIiCIISay m . N t K P C H < H S H
TT-FBP IF-SH
IM V m N G IyA jx^^ ,
3. T T - S H + ll"FEiP —— - ►
b h V
Figure 4. Synthesis scheme of In-Tf conjugate. 1. Synthesis of PDP-
insulin 2. Synthesis of Tf-SH 3. Synthesis of In-Tf.
55
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
The SPDP modified insulin was next coupled to free-sulfhydryl containing Tf (Tf-
SH). Tf-SH was synthesized by modifying Tf with SPDP. Human serum apo-Tf (20
mg/mL in PBS, pH 7.0) was loaded with iron by incubating the protein with ferric
ammonium citrate (10 mg/mL in PBS; added to apo-Tf in a 1:10 ratio of ferric
ammonium citrate : apo-Tf) at 37° C for 2 h. The reaction mixture was then dialyzed
over night to remove excess ferric ammonium citrate. The iron-loaded Tf (holo-Tf)
was then modified with SPDP (1.6 mg in di-methyl formamide) at 4° C for 30 min.
The reaction mixture was then dialyzed overnight against PBS (pH 8.0).
Tf-SH was generated subsequently by reducing PDP-Tf with dithiothreitol (25 mM)
for 15 minutes. Tf-SH was then purified from the excess dithiothreitol by size
exclusion chromatography (Sephadex G-50). The purified Tf-SH was immediately
added to two-fold molar excess PDP-insulin and the reaction mixture was well-
stirred for 2 h at 4° C. The reaction was quenched by adding N-ethyl maleimide to a
final concentration of 1 mg/mL. The conjugate was purified by Sephadex G-50
chromatography in PBS (pH 8.0) mobile phase.
2.10 Cloning and production of G-CSF
Human G-CSF was cloned by RT-PCR from human bladder carcinoma cell line
5637 (ATCC), as described by Souza et al (348) and subsequently sub-cloned into
56
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
the pGEX-4T-l expression vector (Amersham Pharmacia) as a GST fusion protein in
BL21 E. coli. The expression of the fusion protein was accomplished hy growing
the transformed BL21 bacteria in LB broth at 37° C until A600nm = 0.5 followed by
induction of GST-G-CSF synthesis with 0.1 mM isopropyl P-D-thiogalactoside
(IPTG) for four hours. The bacteria were then harvested, resuspended in PBS, and
lysed via sonication. Triton X-100 was added to the sonicate for a final
concentration of 1% and gently mixed on ice for 30 min. GST-G-CSF fusion protein
was purified from the crude sonicate with Glutathione-Sepharose 4B, washed
extensively with PBS, and incubated overnight with 5U/mg of thrombin (Amersham)
to liberate G-CSF from the matrix. The minute amount of thrombin was removed
from the eluent via Benzamidine Sepharose 6B (Amersham) treatment. The purity
of G-CSF was verified via SDS-PAGE and the yield was estimated by measuring the
absorbance at 280nm, with an extinction coefficient of 15820 M'^ The yield of G-
CSF from this procedure was typically 0.5 mg/L.
2.11 Preparation of Tf-G-CSF Conjugate
GCSF (Amgen, Thousand Oaks, CA or alternatively the cloned product that
was synthesized) was covalently linked to iron-loaded human Tf through disulfide
linker chemistry (Figure 5). Briefly, a 20 mg/mL solution of iron-loaded Tf in PBS
(pH 7.4) was mixed with a ten-fold molar excess of the hetero-bifunctional cross-
linking agent A-Succinimidyl 3-(2-pyriyldithio)propionate (SPDP) (Pierce,
57
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Rockford, IL) at 4° C for 30 minutes. The reaction mixture was then dialyzed
overnight against PBS (pH 8.0) to remove the excess SPDP. The final ratio of SPDP
to Tf was determined to be 2:1. GCSF was next modified with the homo-
bifunctional linker agent 1,4-Di- [3 ’ -(2 ’ -pyridyldithio)propionamido]butane
(DPDPB) (Pierce) by targeting the free sulfhydryl group of Cysl7 in GCSF. A ten
fold molar excess of DPDPB was added to a 10 mg/mL solution of GCSF in PBS
(pH 7.4) and allowed to react at room temperature until a maximum absorbance at
343nm was obtained. The DPDPB-modified GCSF was subsequently dialyzed
overnight against PBS (pH 8.0, 4° C) to remove excess linker agent. The ratio of
DPDPB to GCSF was determined to be 1 : 1. S PDP-Tf was reduced with 25 mM
dithiothreitol (Sigma) to generate the reactive sulfhydryl species and reacted with 10-
fold molar excess DPDPB-GCSF to form the GCSF-Tf conjugate with a 22.8
angstrom aliphatic spacer. Excess GCSF was needed to limit the extent of unwanted
Tf-Tf crosslinking. The reaction was quenched by addition of 1 mg/mL of N-
ethylmaleimide, followed by overnight dialysis against PBS (pH 8.0,4° C). The Tf-
GCSF conjugate was purified by gel filtration on a Sephacryl S-200 column.
58
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
A)
Cysl7-SH
+ D PD PB
iCysl7-SS(CH2)2CONH(CH2)2NHCO(CH2)2SSPyr
. ( i. M L
i ^ Cysl7-SS(CH2)2CONH(CH2)2NHCO(CH2)2SSPyr
i
GCSi
N H C 0 (CH2)2SH
Cysl7-SS(CH2)2CONH(CH2)2NHCO(CH2)2SS(CH2)2CO
G-CSF-Tf
Figure 5: Synthesis scheme for G-CSF-Tf. A) G-CSF is modified with
DPDPB by targeting the free sulfhydiyl group of Cys 17. B) DPDPB-modified
G-CSF is combined with reduced SPDP-modified Tf to form G-CSF-Tf.
59
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2.12 Assessment of Apical-to-Basolateral Transcytosis of ^^^I-Tf, ^^®I-In-Tf,
G-CSF and ^^®I-G-CSF-Tf across Caco-2 and MDCK Cell Monolayers
lodinated G-CSF was obtained for the in vitro transport studies by purifying r-
metHu-G-CSF from commercial filgrastim preparations (Amgen). Specifically,
filgrastim solution was dialyzed against lOmM acetate buffer, pH 4.0, to remove
excipients. Following this, the recovered solution was concentrated in Amicon 3,000
MWCO concentrators, and the G-CSF was iodinated by the chloramine-T catalyzed
method. G-CSF and Tf-G-CSF were iodinated using the chloramine-T method as
described above for the preparation of ^^^I-Tf. Transport studies were conducted on
two week-old Caco-2 monolayers, 6 or 7 days after they had exhibited signs of tight
junction development. Monolayers were washed once with DMEM and incubated at
37° C for 45 min to deplete endogenous Tf. Media were subsequently replaced and
the monolayers were treated with ^^^I-Tf, ^^^I-G-CSF or ^^^I-Tf-G-CSF in the apical
compartment (1.5 jig/mL). Non-specific transport was measured in parallel by the
inclusion of 100-fold molar excess of unlabeled Tf. A transcytosis enhancer, i.e,
BFA (1.6 pg/mL), AG-10 (500 pM), wortmannin (1 or 10 pM), or LY294002 (100
mM) was included in the apical media at the beginning of the transport study. At 2,
4, and 6 h post-dosing, 500 pL samples were collected from the basolateral
compartment and replenished with an equal volume of fresh DMEM. Samples were
subjected to 15% trichloroacetic acid (TCA) precipitation and radioactivity of the
pellet was measured with a Packard gamma counter. The extent of TfR-mediated
60
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
transcytosis was determined by subtracting non-specific transport (inclusive of
excess unlabeled Tf) from total transport.
2.13 Assessment of Apical-to-Basolateral Transcytosis of ^^® I-T f, ^^®I-GCSF and
i25i-Tf.GCSF across Rat Alveolar Epithelial Cell Monolayers
GCSF and Tf-GCSF were iodinated using the chloramine-T method as described
above for the preparation of ^^^I-Tf. Transport studies were conducted on day 6 or 7
rat AEG monolayers, with or without KGF treatment from day 0. Monolayers were
washed once with MDSF and incubated at 37° C for 45 minutes to deplete
endogenous Tf. Media were subsequently replaced and the monolayers were treated
with ^^^I-Tf, ^^^I-GCSF or *^^I-Tf-GCSF in the apical compartment (1.5 jxg/mL).
Non-specific transport was measured in parallel by the inclusion of KXTfold molar
excess of unlabeled Tf. A transcytosis enhancer, i.e., BFA (1.0 pg/mL), AG-10 (500
pM), wortmarmin (1 or 10 pM), or LY294002 (100 mM) was included in the apical
media at the beginning of the transport study. At 2,4, and 6 hours post-dosing, 500
pL samples were collected from the basolateral compartment and replenished with
an equal volume of fresh MDSF. Samples were subjected to 15% trichloroacetic
acid (TCA) precipitation and radioactivity of the pellet was measured with a Packard
gamma counter. The extent of TfR-mediated transcytosis was determined by
subtracting non-specific transport (inclusive of excess unlabeled Tf) from total
transport.
61
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2.14 Analysis of Transcytosed Proteins
Transcytosed proteins were analyzed in separate transport studies via size exclusion
chromatography and GCSF-dependent cell proliferation assays. Basolateral fluids
were collected after a six hour transport study as described above and subjected to
Sephacryl S-200 column chromatographic analysis, where stock ^^^I-labeled proteins
(i.e.^^^I-Tf, ^^^I-GCSF and ^^^I-GCSF-Tf) were used for determination of appropriate
elution volumes. Biological activity assays were conducted for transcytosed
GCSF and *^^I-GCSF-Tf by measuring proliferation of the murine myeloblastic cell
line NFS-60 (349) (courtesy of Dr. J. Dile, St. Jude’s Children’s Research Center,
Memphis, TN). NFS-60 cells that had been cultured in RPMI-1640 medium,
supplemented with both 10% FBS and 10% WEHI-3 (ATCC) conditioned medium
(CM), were washed three times with serum- and WEHI-3 CM-free RPMI-1640 and
aliquoted to 96 well microtiter plates at a density of 1 x 10^ cells/mL. These cells
were spiked with 20 )iL of medium that had been previously recovered from the
basolateral compartments in the conjugate transport studies and concentrated 10-fold
with a Centricon centrifugal concentrator apparatus (Amicon, Bedford, MA). The
samples were incubated at 37°C in a 5% CO2 incubator for 48 hours. A MTT (3-
(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was
subsequently performed essentially as previously described by Mosmann (350).
Briefly, cells were incubated with phenol-red free RPMI media (Invitrogen,
62
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Carlsbad, CA) containing 1 mg/mL MTT for two hours. The formazan crystals that
formed were then dissolved in isopropanol (Sigma) and absorbance was measured at
570nm on a Dynatech M70 microplate reader.
2.15 In vivo studies of subcutaneously and orally delivered G-CSF-Tf conjugate
in male BDFl mice
Specific pathogen-free BDFl male mice (Charles River Laboratories, Wilmington,
MA) were used at 6-8 weeks of age. BDFl mice were chosen due to their relatively
robust stimulatory response to human G-CSF. The mice were allowed to acclimate
for several days prior to beginning experiments. Animal experiments were
compliant with the 'Principles of Laboratory Animal Care' (NDH Publication #85-23)
and approved by the lACUC at USC. The mice were fasted for 12 h prior to dosing.
The mice were individually weighed and the dosages were adjusted accordingly.
The mice weight was typically between 18-20 g. The treatment groups (n = 5)
received a single subcutaneous (SC) injection or oral administration (via gavage
needle) on day 0 with G-CSF-Tf, filgrastim, or vehicle control (PBS). Since the
molecular weight of G-CSF-Tf is about 5-fold higher than that of filgrastim, G-CSF-
Tf was administered at 5 mg/kg SC and filgrastim was given at 1 mg/kg SC.
Similarly, G-SCF-Tf was given at a dose of 50 mg/kg orally and filgrastim was given
at 10 mg/kg orally. The concentrations of the dosing solutions were adjusted so the
dosage volume in all cases was 100 pL. G-CSF-Tf and filgrastim were formulated
63
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
with sodium bicarbonate (30 mg/mL) in PBS prior to oral administration. Sodium
bicarbonate was included to neutralize stomach acid in order to prevent hydrolysis of
the protein-drug.
Blood samples were collected daily from the tails of the mice into micro
tubes that had been pretreated with heparin. Total white blood cell counts (WBC)
were performed manually with a hemacytometer. The samples were diluted 20-fold
and lysed in an acidic crystal-violet solution (0.1% crystal violet, 1% acetic acid, in
water) prior to being loading in the hemacytometer. The percentage of poly
morphonuclear neutrophils (FMN) amongst the leucocytes was determined manually
with Wright-stained blood-smear glass slides that were examined with a lOOx oil
immersion lens on an Olympus BH-2 microscope. The absolute number of
neutrophils was determined by applying the observed FMN percentage against the
total WBC count for each sample (351).
2.16 Statistical analyses
Data are presented as mean (±SEM). One-way analysis of variance (ANOVA) was
used to determine statistical significance among group (n > 3) means using Tukey’s
post-hoc tests. p<0.05 was considered significant.
64
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
RESULTS
3.1 Synthesis and characterization of G-CSF-Tf
The G-CSF that was cloned, expressed, and purified from E. coli was analyzed to
verify that the biological activity was maintained through the purification process.
As seen in Figure 6 A, the purified G-CSF closely matched the Neupogen standard in
the ability to stimulate proliferation of NFS-60 cells, a murine cell line that is
specifically stimulated by murine and human G-CSF. In addition when the purified
G-CSF is analyzed in a dose dependant MTT assay, the ED50 appears to closely
match the values that have been reported for Neupogen, with an ED50 of
approximately 0.06 ng/mL (Figure 6 B).
G-CSF-Tf conjugate was synthesized using disulfide DPDPB linker chemistry that
targeted the free sulfhydryl moiety of Cys 17 on G-CSF, while using the
heterobifunctional linker SPDP to create a Tf that contained reactive sulfhydryl
species. The free sulfhydryl containing Tf was formed by a short reducing reaction
under mild DTT solution (25 mM).
SDS-PAGE demonstrated that the GCSF-Tf conjugate was dominated by
cross-hnked species, with the majority of the non-reduced conjugate showing
coomassie blue staining near or above the 205 kDa molecular weight standard band
65
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(Figure 7, lane 1), indicating the presence of oligomers of Tf in the conjugate. While
the band that appears for the conjugate is broad, it does not appear to have significant
amounts of free Tf or G-CSF. However, when the conjugate is reduced by a short
incubation with DTT, the conjugate is cleaved to liberate its two constituent proteins.
As seen in lane two of Figure 7, two bands appear near 80 kDa and 20 kDa, which
coincide with the expected molecular weights of Tf and G-CSF.
The conjugate also demonstrates a distinct elution profile when analyzed by
size exclusion chromatography. For example, when the conjugate was applied to a
40 mL Sephacryl 200 column, the conjugate eluted at fraction 17 while standard
solutions of Tf and G-CSF eluted at fractions 19 and 23 respectively, as shown by
absorbance of the fractions at 280 nm (Figure 8). In addition, when the eluted
fractions are analyzed for ability to stimulate proliferation of NFS-60 cells, the
absorbance at 570 nm displayed a maximum at fraction 17 (Abs570n m = 0.2) and
appeared to coincide with the maxima observed for the conjugate concentration
curve (Abs. at 280 run). The conjugate also appeared to be well separated from un
reacted free G-CSF, with only a minimal amount of cell-proliferation ability
demonstrated at fraction 23 (Abs570n m = 0.04).
66
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 6. G-CSF-dependent proliferation of NFS-60 cells.
(A) NFS-60 cells were seeded at 20,000 cells/mL and treated with
commercially available G-CSF (filgrastim) or G-CSF derived
purified from E.coli through a GST-G-CSF fusion protein construct
at a dose of 1 ng/mL. Cell concentration was determined daily with a
Coulter counter. (B) G-CSF dose dependent assay of NFS-60 cell
prohferation. NFS-60 cells were seeded in 96-well plates at a density
of 10,O C X ) cells/mL. The wells were spiked (in triplicate) with
varying concentrations of G-CSF and incubated at 37 °C in 5% CO2
for 48 hours. The growth media was then replaced with phenol-red
free media containing 1 mg/ml MTT and allowed to incubate for 2
hours. After incubation, the MTT-containing media was replaced
with 2-propanol to dissolve the formazan crystals. Absorbance was
measured at 570 nm.
67
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
45 -
40 -
35 -
-X- Filgrastim
Recombinant GCSF
Control
30 -
« 20 -
15 - I
10
0 24 48 72 96
Culture Time (hrs)
NFS-60 MTT Assay
1.2
h- 0.8
O 0.6
is 0.4
< 0.2
0.000 0.005 0.01 0.02 0.04 0.08 0.16 0.31 0.63 1.25 2.5
Lyophilized G-CSF (ng/mL)
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
205 kDa
• i M 121 kDa
70 kDa
52 kDa
35 kDa
29 kDa
21 kDa
1
Figure?. SDS-PAGE analysis of G-CSF-Tf. G-CSF-
Tf was analyzed under non reducing (lane 1) and
reducing (lane 2) conditions in a 10 % acrylamide gel.
The gel was stained with Coomassie Blue.
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
0.20
0.14
0.18
0.16
E 0.12
GCSF-Tf
0.14
I 0.10
O I
m 0.08
0.12
G -CSF
0.10
GCSF-Tf
A570
0.08
0.06
0
eo
01 0.06
0.04
0.04
J Q
0.02
0.02
0.00 0.00
T -
CO CO
^i ^ o c o c o c r j c MLOo o
T - T - T ” T - CM o a CM
Fraction
Figure 8. Chromatographic analysis of G-CSF-Tf. G-CSF-Tf
was analyzed by S-200 gel chromatography (40 mL column).
The purity of the synthesized G-CSF-Tf was estimated by
comparing the position of eluted G-CSF-Tf to that of G-CSF and
Tf standards applied to the same column by measuring the
absorbance of the eluted samples at 280nm. The bioactivity of
the eluted samples was determined by the ability to stimulate
proliferation of NFS-60 cells as measured by MTT assay (Abs at
570nm).
( Q
m
E
c
o
lO
m
70
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.2 The Effect of Wortmannin on TfR-Mediated Transcytosis in Caco-2
Monolayers
In light of previous findings that have shown that GTPase inhibitors are able
to elicit an enhancement of TfR-mediated transcytosis of protein drugs in vitro and in
vivo (113; 312), efforts were undertaken to examine whether or not inhibiting the
functions of downstream effectors of Rabs would result in a similar enhancement of
TfR-mediated transcytosis. Towards this end, PD kinases were identified as a
potential target for inhibition in order to alter sorting events to favor an increase in
net apical-to-basolateral transport. When the natural PDK inhibitor, wortmannin,
was applied to the apical compartment of Caco-2 monolayers at the beginning of a 6-
hour transport study in concentrations of either 1 or 10 |iM, a significant
enhancement of TfR-mediated transcytosis was observed (Figure 9). Specifically,
the TfR-specific apical-to-basolateral transport rate of ^^^I-Tf was 0.58
ng/hr/monolayer for the 10 pM wortmannin treatment group and 0.49
ng/hr/monolayer for the 1 jiM treatment group while the control monolayers
demonstrated a basal level TfR-mediated ^^^I-Tf transport rate of 0.28
ng/hr/monolayer. The monolayer integrity as determined by TEER measurement,
remained unaffected during the time course of the experiment, with both control and
10 jiM wortmannin treatment groups exhibiting nearly identical resistances over the
course of the experiment (Figure 10).
71
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.5
Wortmannin 10 uM
Wortmannin luM
2.5
Control
05
0.5
0 1 2 3 4 6 5
Time (h)
Figure 9. Wortmannin induced enhancement of TfR-mediated
transcytosis in Caco-2 monolayers. Two-week old Caco-2 monolayers
were dosed with 1.5 |xg/mL of in the apical compartment.
Nonspecific transport was determined parallel by the addition of 100-fold
excess unlabelled Tf. Wortmannin was spiked into the apical bathing
fluid at the beginning of the transport experiment. Samples were
recovered from the basolateral compartment at regular intervals and the
radioactivity of the TCA precipitatable fraction was determined by
gamma counter (n=3).
7 2
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
500
450
s r ^ o o
o
g 300
o 2 5 0
- 200
O J 150
UJ
H 100
Wortmannin 10 uM
Control
6 0 2 4
Time (h)
Figure 10. Wortmannin’s effect on TEER of Caco-2 monolayers.
Two-week Caco-2 monolayers were spiked with wortmannin to a final
concentration of 10 fiM in the apical compartment. The TEER was
measured over a 6-hour time course with a voltohmmeter (n=3).
73
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.3 LY294002 Mediated Enhancement of TfR-mediated Transcytosis in Caco-2
Monolayers
Since wortmannin is a non-specific phosphatidyl inositol kinase inhibitor,
LY294002 was chosen as a agent to be used for the exploration of a specific inhibitor
of PB-K. When LY294002 was applied to the apical compartment of the beginning
of a 6-hour transport study to a final concentration of 100 |iM, a significant
enhancement of TfR-mediated transcytosis was observed (Figure 11 A). Specifically,
the steady state TfR-specific apical-to-basolateral transport rate of ^^^I-Tf was 0.55
ng/hr/monolayer for the 100 jiM LY294002 treatment group while the control
monolayers demonstrated a roughly 5-fold lower basal level TfR-mediated *^^I-Tf
transport rate of 0.11 ng/hr/monolayer. The monolayer integrity as determined by
TEER measurement, remained unaffected during the time course of the experiment,
with both control and 100 jiM LY294002 treatment groups exhibiting nearly
identical resistances over the course of the experiment (Figure 12). In addition, as
can be seen in Figure 1 IB, the increase in ^^^I-Tf transport was due to an
enhancement of TfR-mediated processes, with the nonspecific ^^^I-Tf transport
remaining unchanged in the presence of 100 jiM LY294002 (Figure 5). The rate of
non-TfR-specific *^^I-Tf transport was 0.12 ± 0.015 ng ’ ■^^I-Tf/well/hr for the
LY94002 treatment group and 0.11 ± 0.016 ng ^^^I-Tf/well/hr for the control group.
7 4
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 11. LY294002 induced enhancement of TfR-mediated
transcytosis in Caco-2 monolayers.
Two-week old Caco-2 monolayers were dosed with 1.5 jig/mL of
in the apical compartment. Nonspecific transport was determined
parallel by the addition of 100-fold excess non-radioabelled Tf.
LY294002 was spiked into the apical bathing fluid at the beginning of the
transport experiment. Samples were recovered from the basolateral
compartment at regular intervals and the radioactivity of the TCA
precipitatable fraction was determined by gamma counter. The time
course of TfR-mediated transport for the LY294002 and control groups is
shown in figure A. The average transport rate observed for the total,
nonspecific, TfR-mediated portions of the LY294002 and control groups
over the full six-hour period is shown in figure B (n=3)
75
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.75-
L\»M }Q2100uM
c
Cortro! 1.25 4
r
I
la
CM
"0
^ 0 .7 5 j
Q.
( f >
S 0.5-
c o
h“
0 .2 5 4
1 2 6 0 3
Time(h)
4 5
0.80 - 1
0.70 -
0.60
0.50
h-
DS
S. 0.40
B
1 0.30
■ c
§. 0.20
O T
C
2 0.10
0.00
B
■ LY294002 100 u M
■ Control
Total Non-specific TfR-mediated
7 6
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
700 n
600 i:
£ 500 "-
o
E 400 -
300 -
♦ — LY294002 100 uM
200 -
■ ■ — Control
100 H
0 1 2 3 4 5 6
Time (h)
Figure 12. LY294002’s effect on TEER of Caco-2 monolayers. Two-
week old Caco-2 monolayers were spiked with LY294002 to a final
concentration of 100 jxM in the apical compartment. The TEER was
measured over a 6-hour time course with a voltohmmeter (n=3).
77
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.4 Effect of BFA, AG-10, and LY294002 on TfR-Speclfic Binding in
Caco-2 Monolayers
In order to determine the effect that LY294002 has on TfR distribution, two-
week old Caco-2 monolayers were dosed with 100 mM LY294002 in the apical
compartment, incubated at 37° C for 1 h and then radiolabeled Tf was applied to the
apical or basolateral compartment of the monolayers and the monolayers were
incubated at 4° C for two hours. Likewise, separate Caco-2 monolayers were also
simultaneously dosed with BFA and AG-10 (1.6 pg/mL and 500 mM, respectively)
and ^^^I-Tf was incubated at 4° C for two hours. Non-specific binding of Tf was
determined in parallel. As can be seen in Figure ISA, LY294002 treatment appeared
to result in a significant increase in TfR-specific binding at the apical surface
compared to the BFA and control treatment groups, with 119 ± 11 fmol TfR/
monolayer for the LY294002 treatment group and 68 ± 7 fmol TfR/monolayer and
84 ± 6 fmol TfR/monolayer detected for the BFA and control treatment groups,
respectively.
In contrast, LY294002 treatment resulted in a slight but significant decrease
in TfR levels present at the basolateral surface of Caco-2 monolayers compared to
control with 1920 ± 80 fmol TfR/monolayer and 2100 ± 85 fmol TfR/monolayer for
the LY294002 and control treatment groups, respectively (Figure 13B). In likewise
fashion, AG-10 and BFA treatment resulted in a significant decrease in TfR levels
7 8
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 13. The effect of BFA, AG-10, and LY294002 on the
surface distribution of TfR in Caco-2 monolayers.
TfR levels were determined through TfR specific binding of ^^^I-Tf
(1.5 |ig/mL) to the apical (A) and basolateral (B) surfaces of two-
week old Caco-2 cultures grown on 24mm Trans well filters. Non
specific binding was determined in parallel by the addition of 100-
fold molar excess of non-radiolabeled Tf to the respective donor
fluid. BFA (1.6 |Xg/mL), AG-10 (500 pM), and LY294002 (100
pM) were included in the in apical bathing fluid in all cases and
allowed to incubate for two hours at 37°C prior to replacing the
media with fresh media that had been stabilized at 4°C and which
contained the radiolabeled Tf. The surface binding experiments
were conducted at 4°C for two hours. TfR-specific binding was
determined as the difference between total and non-specific ^^® I-T f
binding. The monolayers were excised after incubation and the
level of radioactivity was determined on a Packard gamma counter.
79
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
> 100
LY 294002 AG-10 BFA Control
0)
>
m
o
c
o
E
P
o
E
2000
1500
1000
500
LY 294002 AG-10 BFA Control
B
8 0
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
at the basolateral surface of the Caco-2 monolayers, with 1900 ± 90 fmol
TfR/monolayer and 1750 ± 95 fmol TfR/monolayer for the AG-10 and BFA
treatment groups, respectively.
3.5 Effect of BFA, AG-10, and LY294002 on TfR-specific ®^Fe-Tf uptake in
Caco-2 monolayers
In order to explore the dynamic effects of the transcytosis enhancers on TfR
cycling events, the TfR-specific uptake of ^^Fe-loaded Tf was investigated in the
presence of transcytosis enhancers. In contrast to uptake of ^^^I-Tf, uptake of ^^Fe-Tf
allows for the recording of cumulative endocytosis events since upon entry into the
acidic environment of the endosome the ferric ion will be released from the
internalized Tf, whereas intemalized ^^^I-Tf will be merely recycled back outside of
the cell. As shown in Figure 14A, the apical TfR-specific ^^Fe-Tf uptake over a 6
hour time-course in Caco-2 monolayers is significantly enhanced by BFA treatment,
with 233 ± 44 fmol Tf/cm^ and 40 ± 33 fmol TFcm^ absorbed for the BFA and
control treatment groups, respectively. In contrast, treatment with AG-10 or
LY294002 did not have a significant effect on TfR-specific ^^Fe-Tf uptake in Caco-2
monolayers, with 5 ± 35 fmol Tf/cm^ and 4 ± 61 fmol Tf/cm^ absorbed for the AG
IO and LY294002 treatment groups, respectively. Basolateral uptake of ^^Fe-Tf in
Caco-2 monolayers displayed a different pattern in response to transcytosis enhancer
treatment compared to the effects that were recorded for the uptake at the apical
81
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 14. The effect of BFA, AG-10, and LY294002 on the uptake
of ®^Fe-Tf in Caco-2 monolayers.
TfR internalization was determined through TfR specific uptake of ^Ve-
Tf (1.5 |xg/mL) from the apical (A) and basolateral (B) surfaces of two-
week old Caco-2 cultures grown on 24mm Transwell filters. Non
specific uptake was determined in parallel by the addition of 100-fold
molar excess of non-radiolabeled Tf to the respective donor fluid. BFA
(1.6 |ig/mL), AG-10 (500 pM), and LY294002 (100 pM) were included
in the in either the apical or basolateral bathing fluid at and allowed to
incubate for five hours at 37°C. TfR-specific uptake was determined as
the difference between total and non-specific ® ® Fe-Tf uptake. The
monolayers were excised after incubation and the level of radioactivity
was determined on a Packard gamma counter.
8 2
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
300-
250-
^ 200-
o
P 150-
o
E 100-
50-
0 -
L i j _
C ontrd AG-10 LY
5000
4500
4000
3500
C M
E
3000
u
2500
o
i
2000
1500
1000
500
0
Control BFA AG-1 0 LY
B
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
surface. For example, all three of the transcytosis enhancers significantly reduced
the basolateral TfR-specific uptake of ^^Fe-Tf. BFA treatment caused the largest
percentage decrease in TfR-specific uptake of ^^Fe-Tf at the basolateral surface of
Caco-2 monolayers, with 41% decrease compared to control (2749 ± 70 fmol
Tf/monolayer and 4684 ± 20 fmol Tf/monolayer absorbed over 6 hours for the BFA
and control treatment groups, respectively). LY294002 and AG-10 treatments
caused a 14% and 32% respective decrease in TfR-specific uptake of ^^Fe-Tf at the
basolateral membrane with 4018 ± 40 fmol Tf/monolayer and 3162 ±73 fmol
Tf/monolayer absorbed for the LY294002 and AG-10 treatment groups, respectively,
during a 6 hour uptake experiment.
3.6 Comparison of Transcytosis Enhancers’ Effect on TfR-Mediated Transport
in Caco-2 Monolayers
In order to make a direct comparison between the effects of the transcytosis
enhancers on TfR-mediated transcytosis, two week old Caco-2 Monolayers were
dosed with 1.6 |ig/mL of BFA, 100 fiM LY294002, and 500 pM AG-10 in the apical
compartments at the beginning of a 6-hour transport study. Non-specific transport
was determined in a similar fashion as previously mentioned. As can be seen in
Figure 15, the steady state TfR-specific transport appeared to be enhanced to a
similar degree in the presence of LY294002 and AG-10, with transport rates of 2.00
± 0.16 ng Tf/hr/well and 2.08 ± 0.44 ng Tf/hr/well observed for the LY294002 and
84
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 15. Comparison of transcytosis enhancer effect on TfR-
mediated transcytosis in Caco-2 monolayers.
Two-week old Caco-2 monolayers were dosed with 1.5 jig/mL of
in the apical compartment. Nonspecific transport was determined in
parallel by the addition of 100-fold excess unlabelled Tf. TfR-specific
transport was determined to be the difference between the total and non
specific transport (A). AG-10 (500 fiM ), LY294002 (100 jiM ), or BFA
(1.6 jXg/mL) were spiked into the apical bathing fluid at the beginning of
the transport experiment. Samples were recovered from the basolateral
compartment at regular intervals and the radioactivity of the TCA
precipitatable fraction was determined by gamma counter. The transport
rates were determined for the total, non-specific, and TfR-mediated
components by linear regression for the steady state transport data (B).
In all cases, the error represents the standard error of the mean as
determined by ANOVA (n=3).
85
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
10
CD
I)
c
H
■e
o
a.
w
c
2
H
0 1 2 4 5 6
^ 4 5 -
§ 4 0 -
|a5-
gao-
ffl 2.5 -
| 2 0 -
r i . 5
«" 1-0
1 0.5^
0.0-
i / ^ 1 0
iO 2 9 « 0 2
■ BFA
B
Trtal Non-qDedfic TfFkmedatecI
86
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
AG-10 treatment groups, respectively. The steady state transport rate observed for
the BFA treatment group was significantly less than LY294002 or AG-10, with an
observed rate of 0.8 ± 0.10 ng Tf/well/hr.
3.7 The effect of LY294002 on TfR-mediated transport of a G-CSF-Tf
conjugate across Caco-2 monolayers
Two-week old Caco-2 monolayers, exhibiting TEER levels of approximately
500 Qcm^, were dosed with 1.5 pg/mL of ^^^I-G-CSF-Tf in the apical compartments
of 6-well Transwells. As shown in Figure 16, monolayers that received ^^^I-G-CSF-
Tf in combination with 100 jiM LY294002 exhibited significantly higher TfR-
mediated transcytosis compared to the monolayers that received only ^^^I-G-CSF-Tf.
For example, after six hours, the amount of transported ^^^I-G-CSF-Tf in the
presence of LY294002 was 4-fold higher than the amount of '^^I-G-CSF-Tf
transported in the absence of LY294002 (8.0 ± 0.9 ng/well ^^^I-G-CSF-Tf with 100
pM LY294002, 1.2 ± 0.7 fmol/well ^^^I-G-CSF). In addition, the transport rate was
also significantly higher, with ^^^I-G-CSF-Tf transported at 1.4 fmol/well-hr'^ and
^^^I-G-CSF transported at 0.3 fmol/well-hr ^ Non-specific ^^^I-G-CSF-Tf in the
presence or absence of LY294002 was significantly lower than TfR-mediated
transcytosis, with nonspecific transport composing 25% and 51% of the total
transport for ^^^I-G-CSF-Tf in the presence and absence of LY294002, respectively
(Figure 16 B).
87
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 16. LY294002 induced enhancement of TfR-mediated
transcytosis of ^^®I-G-CSF-Tf across Caco-2 monolayers.
Two-week old Caco-2 monolayers were dosed with 1.5 |ig/mL of
G-CSF-Tf in the apical compartment. Nonspecific transport was
determined in parallel by the addition of 100-fold excess non
radiolabelled Tf. TfR-specific transport was determined as the
difference between the total and non-specific transport (A). LY294002
(100 |i.M), was spiked into the apical bathing fluid at the beginning of
the transport experiment. Samples were recovered from the basolateral
compartment at regular intervals and the radioactivity of the TCA
precipitatable fraction was determined by gamma counter. The
transport rates were determined for the total, non-specific, and TfR-
mediated components by linear regression for the steady state transport
data (B). In all cases, the error represents the standard error of the
mean as determined by ANOVA (n=3).
88
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1 0 1
Q )
i
c
LY»ia)21(X3uM
u.
m
o
?
■ c
a
m
c
c e
Control
H -
1 0 2 3
Time (h)
4 5 6
3.0
2.5
i 2.0
0)
« 1-5
D C
? 1.0
a.
w
2
0.0
B
OLY294002
■ Control
Total Non-specific TfR-mediated
8 9
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.8 Comparison of the effect of BFA, AG-10, or LY294002 treatment on TfR-
mediated Transcytosis in MDCK monolayers
MDCK was chosen as another model cell line that exhibits a high degree of
polarization of TfR distribution. Previous studies have demonstrated that MDCK
and Caco-2 cells demonstrate differing responses to AG-10 treatment, which has
been postulated to be the result of dissimilar TfR recycling pathways within the cell.
In order to make a direct comparison between the effects of the transcytosis
enhancers on TfR-mediated transcytosis, MDCK monolayers exhibiting TEER
values > 3500 0»cm^ were dosed with 1.6 jig/mL of BFA, 100 pM LY294002, and
500 jiM AG-10 in the apical compartments at the beginning of a 6-hour transport
study. Non-specific transport was determined in a similar fashion as previously
mentioned. As can be seen in Figure 17, the steady state TfR-specific transport
appeared to be enhanced to a similar degree in the presence of LY294002 and AG
IO, with transport rates of 2.00 ± 0.16 ng Tf/hr/well and 2.08 ± 0.44 ng Tf/hr/well
observed for the LY294002 and AG-10 treatment groups, respectively. The steady
state transport rate observed for the BFA treatment group was significantly higher
than LY294002 or AG-10, with an observed rate of 0.8 ± 0.10 ng Tf/well/hr. The
relative degree of transcytosis enhancement, when comparing the effects of BFA
treatment to AG-10 and LY294002, was opposite of that seen for the Caco-2
monolayers. The MDCK monolayer integrity as determined by TEER measurement
(Figure 18), was examined over a 6 hour time period in response to BFA (1.6
90
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
|ig/mL), AG-10 (500 |iM), and LY294(X)2 (100 jiM) treatment in the apical
compartment.
Figure 17. Comparison of transcytosis enhancer effect on TfR-
mediated transcytosis in MDCK monolayers.
MDCK monolayers displaying TEER levels > 3500 0«cm^ were dosed
with 1.5 pg/mL of in the apical compartment. Nonspecific
transport was determined in parallel by the addition of 100-fold excess
unlabelled Tf. TfR-specific transport was determined to be the difference
between the total and non-specific transport (A). AG-10 (500 pM),
LY294002 (100 pM), or BFA (1.6 pg/mL) were spiked into the apical
bathing fluid at the beginning of the transport experiment. Samples were
recovered from the basolateral compartment at regular intervals and the
radioactivity of the TCA precipitatable fraction was determined by
gamma counter. The transport rates were determined for the total, non
specific, and TfR-mediated components by linear regression for the
steady state transport data (B). In all cases, the error represents the
standard error of the mean as determined by ANOVA (n=3).
91
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1 2
1 0 -
BFA
LY294002
T-8
Q .
0 1 2 3 4 5 6
Time(lh)
I 3 .0
5 2.0
t 1.5
e 0.5
■AG-10
□ LY294002
□ BFA
B
Total Non-specific TfR-mediated
92
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
4000
3000 -
*E
g 2000 "
o
Control
BFA 1.6 ug/mL
AG-10 500 uM
LY294002 lOOuM
1000 H
0 1 2 3 4 5 6
Time (h)
Figure 18. Effect of BFA, AG-10, and LY294002 treatment on
TEER of MDCK monolayers, monolayers were spiked with
BFA, AG-10, and LY294002 to a final concentrations of
l.Opg/mL, 500 }iM, and 100 pM, respectively, in the apical
compartment of MDCK transwells. The TEER was measured over
a 6-hour time course with a voltohmmeter (n=3).
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Monolayers that received AG-10 and LY294002 exhibited similar reduction in
TEER over the six hour time course, with both treatment groups exhibiting TEER
values near 1200 Q»cm^ at the end of the 6-hour time course. In contrast, the BFA
treatment group exhibited a significantly different TEER at the end of the six hour
time course, with an average TEER of 2200 ± 240 0®cm^. The control monolayers’
TEER did not decrease over the six hour time period.
3.9 Effect of BFA, AG-10, and LY294002 on TfR-Spedfic ^^® I-Tf Binding in
MDCK Monolayers
MDCK monolayers displaying TEER values near 3500 Q.»c t s ^ were treated
with BFA, AG-10, and LY294002 as described above for the Caco-2 monolayers. As
can be seen in Figure 19, BFA treatment resulted in a significant increase in TfR-
specific binding at the apical surface compared to the AG-10, LY294002, and
control treatment groups, with 210 ± 19 fmol TfR/ monolayer for the BFA
treatment group and 150 ± 10, 146 ± 12, and 145 ± 15 fmol TfR/monolayer detected
for the LY294002, AG-10, and control treatment groups, respectively. In contrast,
BFA treatment resulted in a slight but significant decrease in TfR levels present at
the basolateral surface of MDCK monolayers compared to control with 2400 ± 100
fmol TfR/monolayer and 3050 ±110 fmol TfR/monolayer for the BFA and control
treatment groups, respectively. In likewise fashion, LY294002
9 4
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 19. The effect of BFA, AG-10, and LY294002 on the surface
distribution of TfR in MDCK monolayers.
TfR levels were determined through TfR specific binding of *^^I-Tf (1.5
jig/mL) to the apical (A) and basolateral (B) surfaces of two-week old
MDCK cultures grown on 24mm Transwell filters. Non-specific binding
was determined in parallel by the addition of 100-fold molar excess of non
radiolabeled Tf to the respective donor fluid. BFA (1.6 fig/mL), AG-10
(500 joM), and LY294002 (100 |iM) were included in the in apical bathing
fluid in all cases and allowed to incubate for two hours at 37°C prior to
replacing the media with fresh media that had been stabilized at 4°C and
which contained the radiolabeled Tf. The surface binding experiments
were conducted at 4°C for two hours. TfR-specific binding was
determined as the difference between total and non-specific ^^I-Tf binding.
The monolayers were excised after incubation and the level of radioactivity
was determined on a Packard gamma counter.
95
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
200 -
0 )
>.
£ 150
100
E 50
LY 294002 AG-10 BFA Control
3500 1
3000
0 )
« 2500
O
c
o
E
p
■ g 1000
E
500
2000
1500
i 1
t "
LY 294002 AG-10 BFA Control
B
96
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
and AG-10 treatment did not significantly change TfR levels at the basolateral
surface of the MDCK monolayers, with 3050 ± 120 fmol TfR/monolayer and 3100 ±
97 fmol TfR/monolayer for LY294002 and AG-10 treatment groups, respectively.
3.10 Effect of BFA, AG-10, and LY294002 on TfR-specific ®*Fe-Tf uptake in
MDCK monolayers
TfR-specific uptake of ^^Fe-loaded Tf was investigated in MDCK monolayers in the
presence of transcytosis enhancers as described above for Caco-2 cell monolayers.
As shown in Figure 20, the apical TfR-specific ^®Fe-Tf uptake over a 6 hour time-
course in MDCK monolayers is significantly enhanced by BFA treatment, with 350
± 54 fmol Tf/cm^ and 45 ± 30 fmol Tf/cm^ absorbed for the BFA and control
treatment groups, respectively. In contrast, treatment with AG-10 or LY294002 did
not have a significant effect on TfR-specific ^®Fe-Tf uptake in MDCK monolayers,
with 50 ± 35 fmol Tf/cm^ and 39 ± 61 fmol Tf/cm^ absorbed for the AG-10 and
LY294002 treatment groups, respectively. The basolateral uptake of ^^Fe-Tf in
MDCK monolayers displayed a different pattern in response to transcytosis enhancer
treatment compared to the effects that were recorded for the uptake at the apical
surface. For example, BFA significantly reduced ^^Fe-Tf uptake, while AG-10 and
LY294002 did not significantly affect uptake, as seen at the apical surface. BFA
treatment caused a large decrease in TfR-specific uptake of ^^Fe-Tf at the basolateral
surface of MDCK monolayers, with 35% decrease compared to control (2496 ± 100
97
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 20. The effect of BFA, AG-10, and LY294002 on the uptake of
*®Fe-Tf in MDCK monolayers.
TfR intemalization was determined through TfR specific uptake of ^^Fe-
Tf (1.5 jig/mL) from the apical and basolateral surfaces of two-week old
MDCK cultures grown on 24mm Trans well filters. Non-specific uptake
was determined in parallel by the addition of 100-fold molar excess of
non-radiolabeled Tf to the respective donor fluid. BFA (1.6 pg/mL), AG
IO (500 pM), and LY294002 (100 pM) were included in the in either the
apical or basolateral bathing fluid at and allowed to incubate for five
hours at 37°C. TfR-specific uptake was determined as the difference
between total and non-specific ^^Fe-Tf uptake. The monolayers were
excised after incubation and the level of radioactivity was determined on a
Packard gamma counter.
98
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
300
C4
i 250
o
o
E
200
150
100
L i
C o r tr d B F A A G - 1 0 L Y
4500
4000
3500
3000
o 2500
o 2000
E
1 500
1 000
C ontrol
B
B F A AG-1 0 LY
9 9
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
fmol Tf/monolayer and 4100 ±40 fmol Tf/monolayer absorbed over 6 hours for the
BFA and control treatment groups, respectively).
3.11 Measurement of total surface transferrin receptor in primary cultures of
rat alveolar epithelial cell monolayers
Rat AEC monolayers were dosed with media containing 1.5 jig/mL ^^^I-Tf and
incubated at 4°C for two hours to determine total surface receptor binding. TfR-
specific ^^^I-Tf binding was determined as the difference between total and
nonspecifically bound ^^^I-Tf. The level of TfR at the plasma membrane, as
measured by specific binding of ^^^I-labeled Tf to total cell-surface, decreased from a
maximum of 10.7 (± 2.2) fmol/cm^ on day 1 to 1.49 (± 1.3) (n =6) fmol/cm^ on day 7
in culture (Figure 21). The decrease in total cell-surface TfR coincided with
transdifferentiation from the type II to type I cell-like phenotype, which normally
occurs by day 3-4 in culture (329-332).
100
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
I
o
E
14 1
12 -
10 H
T T 1
0 1 2 3 4 5 6 7
Culture day
Figure 21. Total surface TfR in rat alveolar epithelial cells (AEC)
as a function of day in culture. TfR levels were determined through
TfR specific binding of ^^^I-Tf (1.5 pg/mL) to the apical and basolateral
surfaces of AEC cultures grown on 12mm Trans well filters. Non
specific binding was determined in parallel by the addition of 100-fold
molar excess of non-radiolabeled Tf to the respective donor fluid. The
surface binding experiments were conducted at 4°C for two hours.
Data represent the difference between total and non-specific ^^^I-Tf
binding. In all cases, error bars represent standard error of the mean
(SEM) for n = 6. * indicates p<0.05 and ** indicates p<0.01 compared
to that observed for day 1 AEC as determined by one-way ANOVA.
101
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3,12 Total cellular TfR in rat alveolar epithelial cell monolayers as a function of
culture day
Total cellular TfR (soluble receptor fraction) was next assayed to determine
whether the decrease in surface TfR levels was due to a reduction in TfR protein
synthesis or the sequestration of cellular TfR into an intracellular compartment away
from the cell surface (see Table 1). Total soluble TfR on day 1 was 23.2 (± 1.09)
fmol/cm^ (69.5 (± 3.0) pg TfR/pg protein) and decreased to 0.94 (± 0.98) fmol/cm^
(5.3 (± 2.8) pg TfR/pg protein) by day 7 in culture.
Table 1
Total Cellular TfR (0.1% Triton X-100 Soluble Fraction) in Rat AEC.
pg TfR/ug protein fmol TfR/cm^
Day 1 69.5 (± 3.0) 23.2 (± 1.09)
Day 7 5.3 (±2.8)* 0.94 (±0.98)*
Note: Data are presented as mean (+ SEM) for n=3.
* indicates p<0.01 compared to respective Day 1 values as
determined by ANOVA.
102
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.13 Rat alveolar epithelial cell monolayers exposed to keratinocyte growth
factor during culture retain TfR expression
Alveolar epithelial cell monolayers were grown continuously in the presence
of KGF (10 ng/mL) from day 0 through day 7 in culture. Monolayers that were
exposed to KGF exhibited retention of TfR levels at the cell-surface through day 7 in
culture (9.67 (± 1.12) fmol/cm^), while monolayers without KGF exposure
demonstrated a greatly reduced level of TfR by day 7 in culture (1.23 (± 1.31)
fmol/cm^), as compared to that of day 1 AEC cultures (9.81 (± 1.34) fmol/cm^)
without KGF (Figure 22). Exposure of alveolar epithelial cell monolayers to KGF
did not appreciably affect TEER, with both groups exhibiting resistances of about
2,500 flScm ^ on day 7 in culture (data not shown).
3.14 Confocal microscopy of keratinocyte growth factor treated rat alveolar
epithelial cell monolayers
The effect of KGF exposure on TfR levels in rat AEC monolayers was also
investigated by confocal microscopy. Day 1 monolayers (-KGF) and day 7
monolayers {+!- KGF) were stained with monoclonal antibody OX-26, which is
specific for rat TfR (352). Monolayers were subsequently labeled with FTTC-labeled
secondary antibody and visualized via confocal microscopy. Day 7 monolayers that
had been exposed to KGF retained significant levels of TfR staining, similar to that
seen for day 1 cells (Figure 23). This is in contrast to day 7 monolayers that were not
exposed to KGF, which exhibited very
103
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
T
L _ M
H H l
illiiililM
Day7(»KGF) Day 7(+KGF)
Culture Day
Figure 22. Effect of KGF treatment on TfR expression in
rat alveolar epithelial cell monolayers. Total surface TfR was
determined by the specific binding of *^^I-Tf (1.5 |Xg/mL) with
and without KGF exposure (10 ng/mL) of rat AEC grown on 12
mm Trans well filters for seven days. In all cases, KGF was
added to culture media beginning on day 0 and replaced with
each feeding on days 3 and 6. Error bars represent SEM for n =
6. * indicates p<0.01 as compared to that observed in day 7
(+KGF) AEC as determined by one-way ANOVA.
104
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 23. KGF treatment retains TfR expression in rat
alveolar epithelial cell monolayers.
Rat AEC monolayers examined by confocal microscopy for TfR on
days 1 (-KGF) and 7 (±KGF). Monoclonal antibody OX-26 was
used in conjunction with FTTC-labeled secondary antibody to
specifically localize TfR. TfR staining is more pronounced along
the cell-cell borders (i.e. lateral intercellular regions). Bar equals
10 pm.
105
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Day 1 (-KGF)
Day 7 (-
■ M i l
■ ■ ■ ■ ■ ■
illllli llllil M M w M
Day 7 (+KGF)
jr
106
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
little staining for TfR, equivalent to background levels. The confocal images
obtained from the midsection of the monolayers also revealed that TfR staining in
KGF-exposed monolayers was predominantly localized to the basolateral cellular
plasma membrane.
3.15 Type II cell-like rat alveolar epithelial cell monolayers exhibit polarized
distribution of TfR at the cell surface
Previous studies have determined that many other highly polarized cell lines
that form tight monolayers in culture, such as MDCK and Caco-2 cells, localize
expressed TfR almost exclusively to the basolateral surface (353; 354). When KGF-
treated AEC monolayers were selectively exposed to *^^I-Tf, at either the apical or
basolateral surface on day 7 in culture, *^^I-Tf was preferentially bound to the
basolateral surface (Figure 24). The basolateral surface of these monolayers
exhibited 11.8 (± 0.5) fmol TfR/cm^, while the apical surface exhibited only 1.74 (±
2.4) fmol TfR/cm^ (Fig. 3). TfR levels observed for the apical surface were not
significantly different from zero. The basolateral surface of the monolayers not
exposed to KGF exhibited only 1.4 (± 0.5) fmol TfR/cm^, and the apical surface was
again essentially devoid of TfR, exhibiting only 0.19 (± 2.3) fmol TfR/cm^.
107
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.16 Type II cell-like rat alveolar epithelial cell monolayers exhibit polarized
TfR-specific ® ’Fe-Tf uptake
o
E
12
10
8
2
0
Basolateral
(-KGF)
Apical
(-KGF)
Basolateral Apical
(+KGF) {+KGF)
Figure 24. Rat AEC monolayers tested on day 7 for
polarized distribution of TfR. TfR levels were deteraiined
by TfR-specific binding of ^^^I-Tf (1.5 pg/mL) on either the
apical or basolateral cell membranes of AEC monolayers
(+/- KGF). Error bars represent SEM for n = 6. * indicates
p<0.01 compared to basolateral (-KGF) and ** indicates
p<0.05 compared to basolateral (+KGF), as determined by
one-way ANOVA.
108
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
To gain further insight into the functionality of TfR in alveolar epithelial cell
monolayers, TfR-dependent uptake of ^^Fe was determined. ^^Fe-Tf was prepared
and six hour TfR-dependent uptake experiments were performed, at either the apical
or basolateral surface, using day 7 monolayers (+/- KGF). The KGF-exposed
monolayers demonstrated highly polarized ^^Fe-Tf uptake, with a six hour uptake of
229 (±41) fmol ^^e/cm^ from the basolateral surface (Figure 25). In contrast, KGF-
exposed AEG monolayers showed essentially no TfR-dependent ^® Fe uptake [21 (±
33) fmol ^^Fe/cm^] from the apical surface. Monolayers unexposed to KGF
exhibited no significant ^^Fe uptake activity from either the apical or basolateral
surfaces, with respective six hour TfR-dependent ^^Fe uptakes of 0.90 (± 25.0) fmol
^^Fe/cm^ and 0.94 (± 30.9) fmol ^^Fe/cml
3.17 Effect of BFA, AG-10, and LY294002 on TEER of type II cell-like rat
alveolar epithelial cell monolayers
Treatment of rat AEG monolayers with AG-10 (500|iM) and LY294002 (100
|iM) resulted in a rapid decrease in TEER (Figure 26). By the fourth hour of
treatment with AG-10 and LY294002, the TEER had fallen to the point where the
barrier function of the monolayers had begun to fail. In contrast, treatment of AEG
monolayers with BFA resulted in a decrease in TEER to ~1 kOcm^ by the sixth hour.
While the TEER was significantly decreased upon BFA treatment, barrier function
of the monolayers was maintained for the duration of the experiment. BFA was thus
109
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
250 -
E
200 -
o
a >
JF-
w
150 -
O
li
100 -
50 -
0 -
Basolateral
(-KGF)
Apical Basolateral Apical
(»KGF) (+KGF) (+KGF)
Figure 25. TfR-specific uptake of ^Ve in rat AEC monolayers
on day 7. ^®Fe-Tf (2)i.g/mL) was applied to either the apical or
basolateral cell surfaces of AEC monolayers (+/- KGF) and
incubated at 37°C for six hours. Nonspecific ^^Fe uptake was
determined in parallel by the addition of 100-fold molar excess of
unlabeled holo-Tf. TfR-specific uptake was determined as the
difference between total and non-specific ^^Fe uptake. Error bars
represent SEM for n = 3. * indicates p<0.01 compared to apical
(+KGF) and ** indicates p<0.01 compared to basolateral (+KGF)
as determined by one-way ANOVA.
110
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.50
3.00
Control
BFA
^ 2.50
0
1 2.00
1.50
1.00 -
0.50
0.00
0 2 4 6
T im e (h)
Figure 26. Effect of BFA, AG-10, and LY294002
treatment on TEER of rat type II cell-like alveolar
epithelial cell monolayers. Monolayers were spiked with
BFA, AG-10, and LY294002 to a final concentrations of
1.6pg/mL, 500 pM, and 100 pM, respectively, in the apical
compartment of MDCK transwells. The TEER was measured
over a 6-hour time course with a voltohmmeter (n=6).
I l l
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
further evaluated for its potential to be used as a TfR-dependent transcytosis
enhancer in AEC monolayers of the type-II cell-like phenotype.
3.18 Effects of BFA on distribution and endocytosis of TfR in rat alveolar
epithelial cell monolayers
Day 6 KGF-treated type II cell-like monolayers and non-KGF-treated type-I cell-like
monolayers were treated with BFA (1 pg/mL) for two hours. The cell-surface
distribution of TfR was subsequently detected by ^^^I-Tf binding (1 pg/ml) at 4°C for
two hours. TfR-specific binding was determined by subtracting the non-specific
component as measured by the separate inclusion of 100-fold excess non
radiolabeled Tf.
Non-KGF-treated AEC monolayers, or monolayers displaying the type I cell
like phenotype, did not exhibit significant differences in TfR expression at the apical
or basolateral membranes, compared to control AEC monolayers, in response to
BFA treatment (Figure 27A). TfR-specific ^^^I-Tf binding at the basolateral surface
was 1.3 ± 0.3 and 1.5 ± 0.3 fmol TfR/cm^, with and without BFA treatment,
respectively. TfR-specific *^^I-Tf binding at the apical surface was 0.20 ± 1.9 and
0.19 ± 2.0 fmol TfR/cm^, with and without BFA treatment respectively.
As shown in Figure 27B, KGF-treated type II cell-like monolayers exhibited
markedly increased TfR, although the inclusion of BFA (1 pg/mL) only slightly
altered the static distribution of TfR in KGF-treated monolayers. Specifically, KGF-
112
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
treated monolayers showed a small significant reduction in TfR at the basolateral
membrane upon BFA treatment (10.5 ± 0.5 fmol TfR/cm^) as compared to KGF-
treated monolayers that did not receive BFA (11.8 ± 0.4 fmol TfR/cm^). The apical
membrane of KGF-treated AEC monolayers did not show significant differences in
TfR expression upon BFA treatment, with BFA treated monolayers exhibiting 1.2 ±
2.0 fmol TfR/cm^ and control monolayers exhibiting 1.74 ± 1.9 fmol TfR/cm^.
Since *^^I-Tf binding at 4°C presents only a static view of receptor
distribution, efforts were next undertaken to examine what effect BFA might have on
endocytosis of TfR at the apical and basolateral membranes of type I and tj^e II cell
like monolayers. ^^Fe-loaded Tf was applied to either the apical or basolateral
compartment of day 6 monolayers (± KGF), with or without BFA (1.0 ^ig/mL), and
incubated at 37° C for 5 hours. The use of ^^Fe-Tf enabled endocytosis events to be
measured, unlike the use of ^^^I-Tf, since upon endocytosis of the ^^Fe-Tf-TfR
complex, ^^Fe is known to disassociate from the complex in the acidic endosome and
accumulate inside the cell (292). BFA had no effect on ^^Fe-Tf uptake in non-KGF-
treated monolayers (Figure 27 C). However, BFA significantly altered the extent of
endocytosis in KGF-treated monolayers at the apical and basolateral membranes as
shown in Figure 27D. For example, upon BFA treatment, ^^e accumulation at the
basolateral membrane decreased from 225 ± 25 fmol ^®Fe/cm^ to 175 ± 23 fmol
^^Fe/cm^. In contrast, ^^Fe accumulation at the apical membrane increased upon
113
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 27. The effect BFA on surface TfR expression and ®^Fe-Tf
uptake in rat alveolar epithelial cells (AEC).
TfR levels were determined through TfR specific binding of ^^^I-Tf (1.5
pg/mL) to the apical and basolateral surfaces of AEC cultures grown on
12mm Transwell filters in the presence (A) or absence (B) of KGF (10
ng/mL). Non-specific binding was determined in parallel by the addition
of 100-fold molar excess of non-radiolabeled Tf to the respective donor
fluid. The surface binding experiments were conducted at 4°C for two
hours. TfR-specific binding was determined as the difference between
total and non-specific ^^^I-Tf binding. BFA’s effect on TfR-mediated
uptake was determined via ^^Fe-Tf endocytosis. ^^Fe-Tf (2 pg/mL) was
added to either the apical or basolateral compartment of day 6 AEC
monlayers in the presence (C) or absence (D) of KGF (10 ng/mL). Non
specific uptake was determined in parallel by the addition of 100-fold
molar excess of non-radiolabeled Tf to the respective donor fluid. The
monolayers were excised after a six hour incubation and the level of
radioactivity was determined on a Packard gamma counter. BFA was
added to the apical compartment in all cases (1.0 pg/mL). TfR-specific
uptake was determined as the difference between total and non-specific
^Te uptake. In all cases, error bars represent standard error of the mean
(SEM) for n = 6. * indicates p<0.05 compared to respective non-BFA
treated membrane surfaces as determined by one-way ANOVA.
114
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
8
i F A
0 + 8 F A
Basolateral
Basolateral
115
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 27 continued
2i0
200
M
I -
too
50
. 0
2 9 0 -
2 0 0 -
1
m 1 1 0 "
o
2:
m i l -
1 _ J L
Basolateral
BFA
+ BFA
Basolateral
116
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
BFA treatment to 70 ± 10 fmol ^^Fe/cm^ as compared to a negligible uptake of 5.1 ±
8.2 fmol ^^Fe/cm^ without BFA treatment.
3.19 TfR-mediated apical-to-basolateral Tf transport in BFA-treated type II
cell-like rat alveolar epithelial cell monolayers
Transport studies were performed to determine if BFA treatment was able to
elicit TfR-specific apical-to basolateral transcytosis in type II cell-like AEC
monolayers (i.e., KGF-treated) and type I cell-like AEC monolayers (i.e., treated
with control vehicle). Day 7 AEC monolayers (with or without KGF treatment)
were dosed with 1.5 pg/mL of ’^^I-Tf in the apical compartment. The apical
compartments of the monolayers were subsequently spiked with BFA and incubated
at 37° C. Samples were taken from the basolateral compartments at regular intervals,
subjected to 15% TCA precipitation, and radioactivity was counted on a Packard
gamma counter. Non-specific '^^I-Tf transport was determined in parallel by
inclusion of 150 pg/mL of unlabelled Tf. As shown in Figure 2, KGF-treated
monolayers demonstrated a significant increase in TfR-specific transcytosis in
response to BFA treatment. For example, after six hours, the amount of transported
i^Sf-Tf was 4.4-fold higher in the presence of BFA (0.31 ± 0.3 ng/well ^^^I-Tf with
BFA, 0.07 ± 0.02 ng/well ^^^I-Tf without BFA) (Fig. 28A). BFA treatment did not
result in significant change in non-specific transport (0.06 ± 0.3 ng/well '^^I-Tf with
BFA, 0.05 ± 0.02 ng/well ^^^I-Tf without BFA) (data not shown). In contrast, the
117
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 28. The effects of BFA on transport in rat alveolar
epithelial cell monolayers.
Day 7 AEC monolayers, with (A) or without (B) KGF-treatment, were
dosed with 1.5 |Xg/mL of in the apical compartment. The apical
compartments of the monolayers were subsequently spiked with BFA and
incubated at 37° C. Samples were taken from the basolateral
compartments at regular intervals, subjected to 15% TCA precipitation, and
radioactivity counted on a Packard gamma counter. Non-specific ^^^I-Tf
transport was determined in parallel by the inclusion of 10-fold molar
excess of non-radiolabeled Tf. BFA (1.0 (Xg/mL) significantly enhanced
TfR-mediated apical-to-basal transport in KGF-treated (10 ng/mL) AEC
monolayers, yet had no effect on monolayers not treated with KGF. KGF-
treated cells exhibited a small amount of transport in the absence of BFA.
BFA treatment did not result in significant change in non-specific transport
(0.06 ± 0.3 ng/well ^^^I-Tf with BFA, 0.05 ± 0.02 ng/well ^^^I-Tf without
BFA) (data not shown), n = 6.
118
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
0.35 1
0.30 -
+ BFA
&
0.25 -
-BFA
S 0.20 •
i 0.15 ■
0.10 -
0.05 -
0.00
0 1 2 3
Time (h)
4 5 6
0.35 1
KGF
+ BFA
j .
I
-BFA
•g 0.15 -
^ 0.10 -
c
I
^ 0.05 -
0.00
0 2 3
Tim e (h)
4 5 1 6
119
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
type I cell-like AEC monolayers (no KGF) did not demonstrate significant TfR-
mediated ^^^I-Tf transcytosis in the absence or presence of BFA (Figure 28B).
3.20 TfR-mediated apical-to-basolateral transcytosis of In-Tf conjugate across
type II cell-like rat alveolar epithelial cell monolayers
An insulin-transferrin conjugate was synthesized as previously described
(113). Day 7 AEC monolayers (with or without KGF treatment) were dosed with 1.5
|Xg/mL of ^^^I-In-Tf in the apical compartment. The apical compartments of the
monolayers were subsequently spiked with BFA (1.6 pg/mL) and incubated at 37° C.
Samples were taken from the basolateral compartments at regular intervals, subjected
to 15% TCA precipitation, and radioactivity was counted on a Packard gamma
counter. Non-specific *^I-Tf transport was determined in parallel by inclusion of
150 pg/mL of unlabelled Tf. As shown in Figure 29, KGF-treated monolayers
demonstrated a significant increase in TfR-specific transcytosis in response to BFA
treatment. For example, after six hours, the amount of transported ^^^I-ln-Tf was
4.3-fold higher in the presence of BFA (0.30 ± 0.4 ng/well ^^^I-In-Tf with BFA, 0.07
± 0.02 ng/well ’^^I-In-Tf without BFA) (Fig. 2A). BFA treatment did not result in
significant change in non-specific transport (0.08 ± 0.2 ng/well *^^I-In-Tf with BFA,
0.05 ± 0.03 ng/well ^^^I-In-Tf without BFA) (data not shown).
1 2 0
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
0.35
+ BFA
lir— BFA
™ 0.20
X3
©
I 0.15
I 0.10
0.05
0.00
0 2 4 6
Time (h)
Figure 29. ^^®I-In-Tf transport in rat alveolar epithelial cell
monolayers. Day 7 AEC monolayers were dosed with 1.5 |ig/mL
of ^^I-Tf in the apical compartment. The apical compartments of
the monolayers were subsequently spiked with BFA and incubated
at 37° C. Samples were taken from the basolateral compartments at
regular intervals, subjected to 15% TCA precipitation, and
radioactivity counted on a Packard gamma counter. Non-specific
i25i_Tf transport was determined in parallel by the inclusion of 10-
fold molar excess of non-radiolabeled Tf. BFA (1.0 |Xg/mL)
significantly enhanced TfR-mediated apical-to-basal transport in
KGF-treated (10 ng/mL) AEC monolayers, yet had no effect on
monolayers not treated with KGF. KGF-treated cells exhibited a
small amount of transport in the absence of BFA. n = 6.
.21
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.21 Analysis of transcytosed across type II cell-like rat alveolar
epithelial cell monolayers
Apical compartments of BFA-treated (1.0 pg/mL) type II cell-like AEC monolayers
were dosed with 1.5 pg/mL ^^^I-In-Tf and the basolateral media collected after six
hour incubation at 37° C. Samples were subjected to size exclusion chromatography
analysis
When samples recovered from the basolateral compartments were applied to
a 10 ml Sephadex G-50 column, the major recorded peak coincided with the ^^I-In-
Tf column standard at fraction 4, indicating that the molecular weight of the ^^^I-In-
Tf (recovered post-TfR-mediated transcytosis) was identical to the molecular weight
of the *^^I-In-Tf standard. The extent of degradation appeared to be minor, with
relatively little small-molecule products appearing around fraction 10, accounting for
13% of total radioactivity.
3.22 TfR-Mediated ^^®I-GCSF-Tf transcytosis in type II cell-like rat alveolar
epithelial cell monolayers
*^^I-GCSF-Tf transport studies were performed in KGF-treated day 7 AEC
monolayers. The transport studies were performed in an analogous manner to the
i^Sf-Tf studies, with the apical compartment dosed with 1.5 pg/mL *^^I-GCSF-Tf and
122
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 30. Specific TfR-mediated transport of ^^®I-GCSF-transferrin
conjugate determined on day 7 and comparison to ^^®I-GCSF.
The apical compartments of day 7 type II cell-like monolayers were dosed
with 1.5 p,g/mL of ‘^^I-GCSF-Tf (A) or ^^^I-GCSF (B) and spiked with
BFA (1.0 |Xg/mL) at the beginning of the transport study. Samples were
taken from the basolateral compartments at regular intervals, subjected to
15% TCA precipitation, and radioactivity counted on a Packard gamma
counter. Non-specific ^^^I-Tf transport was determined in parallel by the
inclusion of 10-fold molar excess of non-radiolabeled Tf. BFA (1.0
lig/mL) significantly enhanced TfR-mediated apical-to-basolateral
transport in KGF-treated (10 ng/mL) AEC monolayers. Monolayers that
were not exposed to BFA displayed negligible TfR-mediated transcytosis.
BFA treatment did not significantly affect non-specific ^^^I-GCSF-Tf
transport (0.67 ± 0.3 fmol/well ‘^^I-GCSF-Tf with BFA, 0.61 ± 0.3 ng/well
^^^I-GCSF-Tf without BFA) (data not shown). The apical-to-basolateral
transport of '^^I-GCSF was not affected by the presence of BFA, exhibiting
nearly identical transport profiles with or without BFA. n = 6.
123
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
4 .5
% BFA
3.5
- BFA 3.0
li.
2.0
-II-------
o.o;
1 2 0 4 5 3
Time (h)
6
4.5
4.0
I 3.5
+/BFA
, - BFA
1
■c
a
m
c
0.5
0.0
0 2 5 1 3
Tim e (h)
4 6
124
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
then spiked with BFA (1.0 fig/ml). As shown in Figure 30A, BFA significantly
enhanced TfR-specific apical-to-basolateral transcytosis of ^^^I-GCSF-Tf (3.7 ± 0.68
fmol/well '^^I-GCSF-Tf after six hours), while those monolayers that were not
exposed to BFA displayed negligible TfR-specific ^^^I-GCSF-Tf transcytosis (0.21 ±
0.45 fmol/well ^^^I-GCSF-Tf after six hours). BFA treatment did not significantly
affect the degree of non-specific ^^^I-GCSF-Tf transport (0.67 ± 0.3 fmol/well
GCSF-Tf with BFA, 0.61 ± 0.3 ng/well ^^^I-GCSF-Tf without BFA) (data not
shown).
3.23 Comparison of ^^^I-GCSF-Tf and ^^®I-GCSF apical-to-basolateral
transport in AEC monolayers.
The efficacy of BFA-induced enhancement of ^^^I-GCSF-Tf apical-to-basolateral
transcytosis was compared to the apical-to-basolateral transport of ^^^I-GCSF.
Transport studies were performed as described above, with the apical compartments
of day 7 type II cell-like monolayers dosed with 1.5 |ig/mL of ^^^I-GCSF and spiked
with BFA (1.0 p-g/mL) at the beginning of the transport study. TfR-specific
transport of ^^^I-GCSF-Tf (+ BFA) (Figure 3GB) was significantly greater than that
of the similarly treated ^^^I-GCSF group (+ BFA) (Fig. 3B), with respective
cumulative 6-hour apical-to-basolateral transports of 3.7 ± 0.68 fmol/well ^^I-
GCSF-Tf and 1.2 ±0.11 fmol/well ^^^I-GCSF. The apical-to-basolateral transport of
125
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
^^^I-GCSF was not affected by the presence of BFA, exhibiting nearly identical
transport profiles with or without BFA.
3.24 Analysis of transcytosed ^^®I-GCSF-Tf
Apical compartments of BFA-treated (1.0 jig/mL) type II cell-like AEC monolayers
were dosed with 1.5 |Xg/mL ^^®I-GCSF-Tf and the basolateral media collected after
six hour incubation at 37° C. Samples were subjected to size exclusion
chromatography analysis and assayed for bioactivity by measuring the ability to
stimulate NFS-60 cell proliferation (349).
When samples recovered from the basolateral compartments were applied to
a 40 ml Sephacryl-200 column, the major recorded peak coincided with the ^^^I-
GCSF-Tf column standard at fraction 19, indicating that the molecular weight of the
'^^I-GCSF-Tf (recovered post-TfR-mediated transcytosis) was identical to the
molecular weight of the ^^^I-GCSF-Tf standard (Figure 31). The extent of
degradation appeared to be minor, with relatively little small-molecule products
appearing around fraction 40, accounting for 14% of total radioactivity.
The biological activity of transcytosed conjugate was next determined
through a NFS-60 MTT proliferation assay. Basolateral media was collected as
described above. The samples were then sterile filtered, normalized for GCSF
content, and used as assay substrate. The recovered conjugate displayed relatively
minor proliferative ability, with 0.1 ng/mL GCSF equivalents displaying an
126
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
absorbance (570 nm) of only 0.23 ± 0.5 (Figure 32). However, when the sample is
subjected to reducing conditions prior to the assay (25 mM DTT, 15 min), we find
TfR Specific
GCSF-Tf
Transport
-o-G C SF-T f Standard
Fraction #
Figure 31. Gel filtration chromatogram of TfR-mediated
transcytosed ^^®I-GCSF-Tf. Apical compartments of BFA-treated (1.0
|ig/mL) type II cell-like AEC monolayers were dosed with 1.5 |ig/mL
1-GCSF-Tf and the basolateral media collected after six hour
incubation at 37° C. The basolateral samples were applied to a 40 mL
Sephacryl 200 column, eluted with PBS (pH 7.4), and compared to the
standard ^^^I-GCSF-Tf solution. The fractions (1 mL) were subsequently
counted on a Packard gamma counter.
127
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
- ^ ► - gcsf standard
HD- Reduced Conjugate
Non-reduced
Conjugate
0.000 0.006 0.012 0.025 0.050 0.100 0.200 0.400
GCSF Equivalent (ng/mL)
Figure 32. Evaluation of biological activity of transcytosed
GCSF-transferrin. Apical compartments of BFA-treated (1.0
pg/mL) type II cell-like AEC monolayers were dosed with 1.5
pg/mL ^^^I-GCSF or ^^^I-GCSF-Tf and the basolateral media
collected after six hour incubation at 37° C. Biological activity
assays were conducted for transcytosed ^^^I-GCSF-Tf or reduced
^^^I-GSCF-Tf (25mM DTT treatment for 15 min) by measuring
proliferation of the murine myeloblastic cell line NFS-60 via MTT
assay. Results are also shown for GCSF control (Neupogen).
128
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
that biological activity is recovered and fairly closely matches activity of the GCSF
standard, with 0.1 ng/mL GCSF equivalents displaying an absorbance (570 nm) of
0.95 ± 0.07 and 0.80 ± 0.05 for the GCSF standard and the reduced ^^^I-GCSF-Tf
conjugate, respectively.
3.25 Apical-to-basolateral Transcytosis of G-CSF-Tf and TF across Caco-2
monolayers
Two-week old Caco-2 monolayers, exhibiting TEER levels of approximately 500
Ocm^, were dosed with 1.5 pg/mL of ^^^I-G-CSF-Tf or ^^^I-G-CSF in the apical
compartments of 6-well Transwells. As shown in Figure 1, monolayers that received
G-CSF-Tf exhibited significantly higher TfR-mediated transcytosis compared to the
monolayers that received ^^^I-G-CSF. For example, after six, hours, the amount of
transported ’^^I-G-CSF-Tf was 7.8-fold higher than ^^^I-G-CSF (9.3 ± 0.8 fmol/well
*^^I-G-CSF-Tf, 1.2 ± 0.7 fmol/well ^^^I-G-CSF) (Figure 33). In addition, the
transport rate was also significantly higher, with ^^^I-G-CSF-Tf transported at 1.7
fmol/well-hr'^ and ^^^I-G-CSF transported at 0.3 fmol/well h r'\ Non-specific ^^^I-G-
CSF-Tf and ^^^I-G-CSF transport was significantly lower than TfR-mediated
transcytosis, with nonspecific transport composing 25% and 27% of the total
transport for ^^^I-G-CSF-Tf and '^^I-G-CSF, respectively (data not shown).
129
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
10 1
o
G-CSF-Tf
c
I
e
a.
xs
«
1 C
o
a .
(0
c
s
H
0 1 2 3 4 5 6
Tlme(h|
Figure 33. Specific TfR-mediated transport of ^^®I-GCSF-
transferrin conjugate determined in Caco-2 monolayers and
comparison to ^ I-GCSF . The apical compartments of two
week-old Caco-2 monolayers were dosed with 1.5 pg/mL of
G-CSF-Tf or ^^^I-G-CSF. Samples were taken from the
basolateral compartments at regular intervals, subjected to 15%
TCA precipitation, and radioactivity counted on a Packard
gamma counter. Non-specific ^^^I-Tf transport was determined
in parallel by the inclusion of 10-fold molar excess of non-
radiolabeled Tf. The apical-to-basolateral transport of ^^^I-GCSF
was not affected by the presence of excess Tf while ^^^I-G-CSF-
Tf exhibited an 80% reduction in transport (data not shown) n=3.
130
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3.26 Chromatographic analysis of transcytosed G-CSF-Tf
Apical compartments of Caco-2 monolayers were dosed with 1.5 )Xg/mL ^^^I-G-CSF-
Tf and the basolateral media collected after six hour incubation at 37° C. Samples
were subjected to size exclusion chromatography analysis. When samples recovered
from the basolateral compartments were applied to a 40 ml Sephacryl-200 column,
the major recorded peak coincided with the ^^^I-G-CSF-Tf column standard at
fraction 19, indicating that the molecular weight of the ^^^I-G-CSF-Tf (recovered
post-T fR-mediated transcytosis) was identical to the molecular weight of the ^^^I-G-
CSF-Tf standard (Figure 34). The extent of degradation appeared to be minor, with
relatively little small-molecule products appearing around fraction 40, accounting for
13% of total radioactivity.
3.27 Analysis of biological activity of transcytosed ^^I-G-CSF-Tf in Caco-2
monolayers
The biological activity of transcytosed conjugate was next determined through a
NFS-60 MTT proliferation assay (Figure 35). Basolateral media was collected as
described above after a six-hour transport experiment across Caco-2 monolayers.
The samples were then sterile filtered, normalized for G-CSF content, and used as
assay substrate. The sample was subjected to reducing conditions prior to the assay
131
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(25 mM DTT, 15 min) in order to mimic reducing conditions that would encountered
by the conjugate in vivo. The biological activity of the recovered ^^^I-G-CSF-TF
400
350 -i
300 -
-■-TfR Specific G-CSF-Tf
Transport
-o-G -C SF-Tf Standdard
250 i
1 200 -
a.
u
150 -
100
o
Fraction #
Figure 34. Gel filtration chromatogram of TfR-mediated
transcytosed ^^®I-GCSF-Tf. Apical compartments of Caco-2
monolayers were dosed with 1.5 |ig/mL ^^^I-GCSF-Tf and the
basolateral media collected after six hour incubation at 37° C.
The basolateral samples were applied to a 40 mL Sephacryl 200
column, eluted with PBS (pH 7.4), and compared to the standard
'^^I-GCSF-Tf solution. The fractions (1 mL) were subsequently
counted on a Packard gamma counter.
132
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
1.2
G-CSF Startdard
0.8
E
c -o-G-CSF-Tf
e
w 0.6
0.4
0.2
0.0
0.000 0.006 0.012 0.025 0.050 0.100 0.200 0.400
G-CSF Equivalent (ng/mL)
Figure 35. Evaluation of biological activity of transcytosed
G-CSF-Tf. Apical compartments of Caco-2 monolayers were
dosed with 1.5 |Xg/mL ^^^I-G-CSF or ^^^I-G-CSF-Tf and the
basolateral media collected after six hour incubation at 37° C.
Biological activity assays were conducted for transcytosed ^^I-
GSCF-Tf after reduction of the disulfide linkage (25mM DTT
treatment for 15 min) by measuring proliferation of the murine
myeloblastic cell line NFS-60 via MTT assay. Results are also
shown for GCSF control (filgrastim).
133
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
conjugate closely matches the activity of the G-CSF standard, with 0.1 ng/mL G-
CSF equivalents displaying an absorbance (570 nm) of 0.87 ± 0.08 and 0.78 ± 0.06
for the G-CSF standard and the reduced ^^^I-G-CSF-Tf conjugate, respectively.
3.28 Neutrophil proliferation in BDFl mice dosed with subcutaneous and oral
G-CSF-Tf
BDFl mice were given 1 mg/kg filgrastim, 5 mg/kg G-CSF-Tf, or control vehicle
subcutaneously. The day of dosage administration was denoted as day 0. By day 1,
both the G-CSF-Tf and filgrastim treatment groups exhibited an increase in absolute
neutrophil counts (2420 ± 450 cells/|jL for the G-CSF-Tf treatment group and 2375
± 400 cell/|iL for the filgrastim treatment group) (Figure 36). However, by day 3
there was a marked significant difference between the two treatment groups, with
4100 ± 510 cells/fiL for the G-CSF-Tf treatment group and 1200 ± 385 cells/jiL for
the filgrastim treatment groups. The neutrophil levels remained elevated for the G-
CSF-Tf treatment group, relative to control and filgrastim groups, until day 3 and
then returned to normal levels by day 4.
For the oral dosing experiments, BDFl mice were given 10 mg/kg filgrastim,
50 mg/kg G-CSF-Tf, or control vehicle by gavage needle. The mice that received G-
CSF-Tf demonstrated a statistically significant elevation in absolute neutrophil
counts by day 1 with 2350 ± 225 cells/|iL (Figure 37). In contrast, oral
administration of filgrastim did not result in a statistically significant change in
134
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
neutrophil levels compared to control (1080 ± 280 cells/|iL and 990 ± 95 cells/jxL for
filgrastim and control groups on day 1, respectively). Absolute neutrophil counts
continued to be significantly elevated for the G-CSF-Tf treatment compared to
control until day 3, when they retumed to normal levels.
A dose response experiment was next performed on orally administered G-
CSF-Tf. BDFl mice were given either 50 mg/kg, 25 mg/kg, or 12.5 mg/kg G-CSF-
Tf orally by gavage needle. The group that received 50 mg/kg demonstrated a
similar response as seen in the previous experiment, with 2300 ± 295 ceUs/|iL
recorded for absolute neutrophil counts on day 1 (Fig. 6), which was significantly
different from the other two treatment groups. There was an indication that the
neutrophil counts were elevated for the 25 mg/kg group compared to the 12.5 mg/kg
group on day 1 (1400 ± 200 cells/pL and 1030 ± 168 cells/jiL for 25 mg/kg and 12.5
mg/kg groups, respectively, p < 0.10).
135
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
5000
_ 4500
"i
1 4000
"5
— 3500
I
I sooo
0
1 2500
a.
0
Z
2000
1500
1 1000
XI
<
500
* G-CSF-Tf
-♦-Filgrastim
Control
Day 0 Day 1 Day 2 Day 3 Day 4 Day 5
Figure 36. Myelopoietic effect of subcutaneously
administered G-CSF-Tf. G-CSF-Tf (5 mg/kg), filgrastim
(1 mg/mL), or control vehicle was administered
subcutaneously to 8 week old male BDFl mice. Absolute
neutrophil counts were determined daily. * indicates p <
0.01 compared to control.
136
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2500
i
§ 2000
O 1500
Ic
a
z
I
o
< n
XI
<
1000
500
G-CSF"Tf
Filgrastim
Control
Day 0 Day 1 Day 2 Day 3 Day 4 Day 5
Figure 37. Myelopoietic effect of orally administered G-
CSF-Tf. G-CSF-Tf (50 mg/kg), filgrastim (10 mg/kg), or
control vehicle was administered orally by gavage needle to
8 week old male BDFl mice. Absolute neutrophil counts
were determined daily. * indicates p < 0.01 compared to
control.
137
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
2500
2000
■ 4
tn
c
O 1500
a.
8
S 1000
I
0
©
m
§
500
-♦"G-CSF-Tf ( » n ^ )
-■-G-CSF-Tf (25ni/kg)
G^SF-Tf (125 nr^g)
DayO Dayl E :te y 2
Figure 38. Dose response effect of orally administered
G-CSF-Tf. G-CSF-Tf was given orally to 8 week old
BDFl mice by gavage needle at doses of 50 mg/kg, 25
mg/kg, and 12.5 mg/kg. Myelopoietic effect was
determined by daily absolute neutrophil counts. * indicates
p < 0.01 compared to 12.5 mg/kg group.
138
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
DISCUSSION
As shown in Figures 6-8, the G-CSF that was obtained through the GST-G-
CSF fusion construct closely matched the commercially obtained G-CSF (filgrastim)
in the ability to stimulate the proliferation of the G-CSF-dependent murine cell-line
NFS-60. Synthesis of the G-CSF-Tf conjugate was initially attempted using the
heterobifunctional linker SPDP to create a sulfhydryl-reactive modified-Tf that
would be targeted to the free sulfhydryl group of Cysl7 in G-CSF. Even though the
free sulfhydryl group of G-CSF demonstrated reactivity when incubated with
Ellman’s reagent, initial attempts at reacting Tf-PDP with G-CSF-Cysl7 proved
unsuccessful (data not shown). In order to understand why the reaction was not
occurring, the crystal structure of G-CSF was examined using the molecular
visualization computer program, Rasmol. From the crystal structure (Figure 39), it
was apparent that the reaction of SPD-Tf with G-CSF-Cysl7 may have been
impeded due to the fact that Cysl7 of G-CSF resides within a pocket on the surface
of the molecule, thereby suggesting the possibility of steric hindrance to the reaction.
In light of this possibility, a longer -SH reactive linking agent was first used to
modify the free sulfhydryl group so that the -SH reactive species of DPDPB may
reach beyond the borders of the pocket to freely interact with the free -SH groups of
the DTT-reduced Tf-PDP. This modification allowed the reaction to proceed and the
release of pyridine-2-thione was followed by measuring the absorbance at 343nm.
139
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Figure 39. Spacefill model of G-CSF x-ray crystal structure. PDB
coordinates were obtained from the Brookhaven protein data bank and the
image was rendered with Rasmol. The arrow denotes the position of the free
sulfhydryl group of Cysl7, shown in the lighter shaded atom, which resides in
a pocket on the surface of G-CSF.
140
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
G-CSF was conjugated to Tf by disulfide linkage chemistry. Disulfide
linkage chemistry provides the advantage of allowing cleavage of the conjugate to
liberate the free protein-dmg into the circulation (355). Densitometric scanning
analysis of the conjugate revealed that the ratio of Tf to G-CSF in the conjugate was
approximately 1:1 (Figure 7). However, when the non-reduced conjugate was
observed under non-reducing conditions, it appeared to be composed of a
heterogeneous mixture of higher ordered conjugate species (100 kDA to 300 kDa).
It is possible that crosslinking of Tf molecules may occur, as there is multiple
reactive free sulfhydryl groups on the surface of Tf after Tf-PDP has been reduced
with DTT.
Even though the limitations of the current disulfide linkage prevents the
formation of a defined product, the fact that cross-linking occurs between Tf
molecules allows the conjugate to be conveniently separated from unreacted
constituent proteins (i.e. G-CSF and Tf) merely by using size exclusion
chromatography of limited resolution power (i.e. Sephacryl 200) (Figure 8). In
addition, this purification process results in the NFS-60 cell proliferative ability to be
mostly restricted to the protein fraction that contains the conjugate, indicating that
the conjugate is well isolated from its constituent proteins.
Previous findings have demonstrated that the GTPase inhibitor, AG-10, was
able to elicit a dose-dependent TfR-specific enhancement of Tf and In-Tf
transcytosis across Caco-2 monolayers, as well as enhancing the hypoglycemic effect
of orally administered In-Tf in streptozocin-induced diabetic rat (113; 312). Even
141
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
though evidence exists of in vitro and in vivo TfR-specific transport across epithelial
barriers in the absence of enhancers, investigation of agents such as AG-10 is
interesting because uptake of Tf-protein drug conjugates from the GI epithelium is
still relatively inefficient when compared to invasive delivery methods. The rate of
apical-to-basolateral TfR transcytosis can be thought of as relatively low when one
considers that most of the TfR that is present on the surface of polarized intestinal
epithelial cells exists at the basolateral surface of the cell (356). Because of this
polarity, most of the TfR that enters the cell will be from basolaterally derived
endocytosis (357). Alteration of these events, such that the equilibrium of TfR
endocytosis is shifted to some extent toward the apical membrane, could conceivably
result in significant improvement of net apical-to-basolateral flux of Tf-protein-drug
conjugates across epithelial barriers.
As mentioned previously, PD kinases are thought to be downstream effectors
of Rab GTPases. It was postulated that inhibition of PDK could enable a more
specific enhancement of TfR-mediated transcytosis compared to AG-10 treatment.
Initial efforts focused on the effects of wortmannin on TfR-mediated transcytosis of
i^Sf-Tf across Caco-2 monolayers. As was shown in Figure 9, treatment with
wortmannin exhibited a dose dependent and TfR-specific enhancement of *^^I-Tf
apical-to-basolateral transport. In addition, use of wortmannin did not adversely
affect TEER of the monolayers over the course of the experiment (Figure 10),
indicating that integrity of the monolayer’s barrier function was maintained. Though
it is very potent PIK inhibitor, wortmannin is a natural product and it is not very
142
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
specific, affecting a broad range of PIK subtypes (224-227). Because of this,
LY294002 was chosen as a specific inhibitor of PDK to investigate whether
inhibition of events that are downstream of Rab5 functions will result in an
enhancement of TfR-mediated transcytosis that is similar in magnitude or perhaps
even better than that seen for AG-10. As was shown in figure 11-A, LY294002
treatment results in a significant enhancement of total ^^^I-Tf apical-to-basolateral
transport across Caco-2 monolayers. When the ^^^I-Tf transport is examined by
comparing non-specific and TfR-mediated processes (Figure 1 IB), it can be seen
that the enhancement of ^^^I-Tf transport in the presence of LY294002 (100 |iM) is
due to an increase in only the TfR-mediated ^^^I-Tf apical-to-basolateral transport.
LY294002 treatment had no effect on the non-specific transport of ^^^I-Tf across the
Caco-2 monolayers, with both the LY294002 and control groups exhibiting identical
non-specific ^^^I-Tf transport rates. From this data and the fact that LY294002 did
not adversely affect the TEER of Caco-2 monolayers (Figure 12), it is apparent that
treatment with LY294002 does not result in loss of monolayer integrity.
Once it had been established that a PDK inhibitor (i.e. LY294002) was able
to elicit specific enhancement of TfR-mediated transcytosis, efforts were undertaken
next to compare the effect of LY294002 on TfR surface distribution and TfR specific
uptake of ^^e-Tf with that of the previously investigated transcytosis enhancers,
BFA and AG-10. As previously discussed, AG-10 and LY294002 treatment resulted
in a significant increase in TfR at the apical surface of the monolayer (Figure 13). In
contrast, BFA treatment resulted in a significant decrease in TfR at the apical
143
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
surface. When the basolateral membrane was examined, the amount of TfR present
at the surface was not significantly different from control for the LY294002 and AG
IO treatment groups, while the BFA group exhibited a slight but significant decrease
in basolateral surface TfR compared to control. Since BFA treatment has been
shown to result in intemalization and localization of TfR to the trans-Golgi network
(307), one might expect to see a reduction in total surface TfR.
The results from the surface binding studies in Caco-2 cell monolayers in the
presence of transcytosis enhancers would suggest that BFA is acting in a distinct
fashion when compared to AG-10 and LY294002. This difference becomes even
more striking when one looks at intemalization events as opposed to static binding of
^25i-Tf at the cell surface. Uptake studies with ^^Fe-Tf allow for the recording of TfR
intemalization since upon endocytosis, ^Ve will be released from Tf and
accumulated in the cytoplasm as the endosome is acidified. As shown in Figure 14,
BFA treatment had a dramatic and significant effect on Tf uptake from the apical
surface, with an approximately 5-fold increase in ^^Fe-Tf uptake. However, AG-10
and LY294002 apparently did not affect the uptake of ^^Fe from the apical surface,
with results that were not significantly different than control. This result seemingly
contrasts with the ^^^I-Tf transport data across Caco-2 monolayers in the presence
the transcytosis enhancers (Figure 15). As was shown, AG-10 and LY294002 had a
similar and significantly greater enhancement effect on Tf A-to-B transport
compared to BFA. Thus LY294002 would seem to affect a similar TfR recycling
pathway as AG-10.
144
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
This trend is also seen when one examines the effect of LY294002 on TfR-
mediated transcytosis across MDCK monolayers. As shown in Figure 17, BFA had
a significantly greater effect on TfR-mediated transcytosis compared to those
monolayers that received AG-10 or LY294002. In addition, LY294G02 had an
almost identical effect on MDCK TEER as compared to AG-10 over the course of
the transport experiment (Figure 18). The similarity of LY294002 and AG-10
effects on TfR mediated processes is not limited to transport phenomenon.
LY294002 also demonstrates a lack of effect on TfR surface distribution in MDCK
monolayers (Figure 19), as well as ^^Fe-Tf uptake (Figure 20).
Variability in sorting mechanisms for apically destined TfR in Caco-2 and
MDCK cultures forms the basis for understanding the functional differences between
the transcytosis enhancers examined in this study. The predominant mechanisms
whereby cells maintain membrane TfR polarity lies in the ability to sort apical or
basolateral proteins and target them to their respective membranes. There exists two
types of mechanisms by which epithelial cells separate apically destined surface
proteins from those proteins that are meant to be sorted to the basolateral membrane,
namely indirect or direct sorting. This sorting is primarily initiated at two distinct
cellular domains, the trans-Golgi network and the basolateral endosome (358).
Direct sorting, which is used by MDCK cells, utilizes the trans-Golgi network to
fully separate apical and basolateral proteins from each other and deliver these
proteins in a vectorial fashion directly to their respective cellular surface (359). In
addition to direct sorting, Caco-2 cells may utilize indirect sorting of apically
145
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
destined plasma membrane proteins (360; 361). For example, proteins that are
targeting to the apical membrane may initially be directed to the basolateral surface
from the trans-Golgi network. These proteins are then sorted only after they are
endocytosed into the basolateral endosomal system. Once in the basolateral
endosome, the apical proteins are separated from proteins that are destined for
recycling back to the basolateral membrane or for degradation in the lysosomal
compartments, and then transcytosed to the apical surface.
From the data presented in this report, it is postulated that LY294002
dependent enhancement of TfR mediated transcytosis is derived from inhibition of
PD-K dependent processes in the indirect basolateral endosomal sorting of TfR that
is distinct from enhancement by modification of direct sorting mechanisms, as with
BFA (Figures 40 and 41). For example, whereas BFA caused an enhancement of
TfR-mediated transcytosis in both MDCK and Caco-2 cultures, LY294002 treatment
was only able to elicit an enhancement of TfR-mediated transcytosis in Caco-2
cultures (Figure 14). In addition, similarly to previous observations made in human
hepatoma Hep 0 2 cells (362), BFA treatment was shown to decrease TfR expression
at the apical and basolateral surfaces of the Caco-2 cultures and MDCK cultures
(Figures 13 and 19, respectively).
146
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
/Y y yYVYYYVYYYYYYVYYVY|
Direct Sorting
+BFA
Indirect
Sorting
+AG-10
+LY294002
Newly Synthesized
TfR
Figure 40. Possible mechanisms of transcytosis enhancer’s effect
on TfR distribution in polarized epithelia. BFA is known to cause
fusion of TGN and endosomal system and may disrupt the direct
sorting of TfR in MDCK and Caco-2 cells. AG-10, Wortmannin, and
LY2940Q2 may affect Rab and PDK dependent processes in
basolaterally derived TfR sorting. This would result in enhancement of
indirect TfR sorting in Caco-2 cells, but not MDCK cells.
147
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Apical.membrane
Recycling
/
Late ^
endosomes
'X .
C > Recycling
i-L & A iy ^
\
\
/
Basolateral
endocytosis
i
1 FYVL , ___
Rab5
-------- ^
PI-3K
t
AG-10 LY294002
W ortm annin
Figure 41. PI3K Inhibitors as Potential TfR-Mediated
Transcytosis Enhancers. A possible method of action for wortmannin
and LY294002 on TfR distribution in caco-2 cells that may involve
effects downstream from AG-10 activity. LY294002 and wortmannin
may inhibit TfR recycling to the basolateral membrane and result in a
transient shift to the apical plasma membrane .
148
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
X). In contrast, both AG-10 and LY294002 caused an increase in apical surface
expression of TfR and a possible decrease in basolateral expression of TfR in Caco-2
monolayers, while causing no statistically significant change in TfR expression in
MDCK monolayers.
LY294002 treatment of Caco-2 monolayers resulted in an enhancement of
TfR-mediated transcytosis in the apical to basolateral direction, on the other hand the
uptake of ^®Fe-Tf was not enhanced in the presence of LY294002 nor AG-10, while
BFA treatment resulted in a significant increase in ^^Fe-Tf uptake from the apical
membrane. It is possible that since BFA treatment results in the retrograde
movement of TfR to the TGN, that this indirect transcytotic movement of TfR would
allow sufficient time for the acidification of the intravesicular milieu such that ^^Fe
would be released from the TfTfR complex. LY294002 and AG-10 may enhance
the movement of rapid transcellular vesicle trafficking where the vesicles may not be
sufficiently acidified to release ^^e before the vesicle is merges with the plasma
membrane for exocytosis. Or alternatively, the presence of AG-10 and LY294002 in
these cells types may potentially cause a lack of divalent metal ion transporters at the
apical surface, resulting in retention of ^^Fe as the vesicle is transcytosed across the
cell.
Investigations were also performed in primary culture system using rat
alveolar epithelial cell monolayers. The level of TfR at the plasma membrane, as
measured by specific binding of ^^I-labeled Tf to total cell-surface, decreased from a
maximum on day 1 to a minimum on day 7 in culture (Figure 21). The decrease in
149
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
total cell-surface TfR coincided with transdifferentiation from the type II to type I
cell-like phenotype, which normally occurs by day 3-4 in culture (329-332). This
suggests that TfR synthesis may be down-regulated in rat alveolar epithelial cells
upon transdifferentiation to the type I cell-like phenotype and that TfR synthesis
might be limited to alveolar epithelial cells of the type II phenotype.
Total cellular TfR (soluble receptor fraction) was also assayed to determine
whether the decrease in surface TfR levels was due to a reduction in TfR protein
synthesis or the sequestration of cellular TfR into an intracellular compartment away
from the cell surface (see Table 1). The fact that the total soluble receptor also
decreased confirms the decreased synthesis of TfR after ceUs transdifferentiated
towards the alveolar type I cell-like phenotype.
The high level of TfR in cells of the type II cell-like phenotype early in
culture presents a viable target for TfR-mediated transcytosis. However, the lack of
substantial TEER early in culture (e.g., days 0-2) precludes conducting transport
studies using type II cell-like monolayers. Exposure of rat alveolar cell monolayers
to KGF has previously been implicated in prevention of the transition of alveolar cell
monolayers from the type II cell-like to the type I cell-like phenotype. In particular,
KGF exposure of AEG monolayers leads to retention of expression of lung surfactant
proteins and lamellar bodies, which are identifying characteristics of the type H cell
phenotype (333; 336), and KGF prevention and reversal of expression of the type I
cell-specific marker proteins, T la and aquaporin-5 (333; 334).
150
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Alveolar epithelial cell monolayers were grown continuously in the presence
of KGF (10 ng/mL) from day 0 through day 7 in culture. Monolayers that were
exposed to KGF exhibited retention of TfR levels at the cell-surface through day 7 in
culture, while monolayers without KGF exposure demonstrated a greatly reduced
level of TfR by day 7 in culture.
Previous studies have determined that many other highly polarized cell lines that
form tight monolayers in culture, such as MDCK and Caco-2 cells, localize
expressed TfR almost exclusively to the basolateral surface (353; 354). When KGF-
treated AEC monolayers were selectively exposed to ^^^I-Tf, at either the apical or
basolateral surface on day 7 in culture, ^^^I-Tf was preferentially bound to the
basolateral surface. TfR levels observed for the apical surface were not significantly
different from zero, allowing us to conclude that the apical surface can be considered
to be predominantly lacking in TfR. The binding studies for day 7 monolayers not
exposed to KGF support the findings noted above that the amount of TfR present at
the cell surface is greatly reduced in type I cell-like monolayers.
The confocal images obtained from staining with the monoclonal anti-body
OX-26 supported the binding studies that were performed with radiolabeled Tf
(Figure 23). For example, monolayers of the type II cell-like phenotype retained TfR
expression while monolayers of the type I cell-like phenotype did not demonstrate
any TfR-specific staining. The confocal images obtained from the midsection of the
monolayers also revealed that TfR staining in KGF-exposed monolayers was
predominantly localized to the basolateral cellular plasma membrane. This is
151
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
consistent with the normal cellular routing of internalized TfR, where endocytosed
TfR is promptly recycled back to the plasma membrane after the release of ferric
ions in acidic endosomes, thus localizing the potential staining to the basolateral
plasma membrane of the cell at steady state (324).
TfR-dependent uptake of ^^Fe was determined in order to gain insight into the
effect apparent cellular phenotype, i.e. type I or type It cell-like monolayers, had on
TfR endocytosis function (Figure 25). To gain further insight into the functionality
of TfR in alveolar epithelial cell monolayers. The KGF-exposed monolayers
demonstrated highly polarized uptake with most of the TfR-dependent uptake
occurring from the basolateral surface. In contrast, KGF-exposed AEC monolayers
showed essentially no TfR-dependent ^^Fe uptake from the apical surface.
Monolayers unexposed to KGF exhibited no significant ® ^F e uptake activity from
either the apical or basolateral surfaces. The amount of ^^Fe uptake is directly
proportional to the amount of ^^Fe-Tf that has been endocytosed by the monolayers.
^^Fe-free apo-Tf is subsequently recycled to the basolateral surface and exocytosed
by the monolayers (363). Since the molar ratio of ^^Fe:Tf is 2:1, it can be estimated
that the turnover of Tf by AEC monolayers in a six hour period is about 115 fmol
Tf/cm^, which is approximately 10 fold higher than the molar amount of TfR that is
resident on the basolateral surface of KGF-exposed AEC monolayers as determined
by the ^^^I-Tf binding studies (11.8 (± 0.5) fmol TfR/ cm^). This indicates an
apparent half-life for TfR residence at the basolateral membrane of approximately 18
min. These data provide evidence that the elevated synthesis of TfR, as a result of
152
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
KGF exposure, leads to avid ^^Fe uptake activity, and that the activity is restricted to
the basolateral surface.
This part of the study demonstrated that alveolar type Tl-like epithelial cells
express significant TfR, and that synthesis of TfR is down regulated upon
transdifferentiation from the type II to the type I cell-like phenotype. Exposure of
the AEC cultures to KGF results in retention of TfR, indicating that maintenance of
type n cell-like phenotype results in retention of TfR synthesis. In addition, the
synthesized TfR is limited exclusively to the basolateral surface of AEC monolayers
and TfR activity, as determined by TfR-specific ^^Fe-Tf uptake, is restricted
predominantly to the basolateral surface of KGF-exposed AEC monolayers. The
presence of TfR exclusively at the basolateral surface of AEC monolayers exhibiting
type n cell-like characteristics indicates that type II cells are likely involved in the
transcytotic transport of Tf between plasma and alveolar lining fluid. These findings
may provide insight into how Tf, an important constituent of the surface fluid lining
the alveoli of the lung, is present at relatively high concentrations in alveolar fluid.
Elucidating the magnitude, directionality, and regulation of these putative processes
presents an attractive area for future research.
We previously reported that Tf-protein drug conjugates have been
successfully used to facilitate the transcytosis of the protein drugs across epithelial
barriers in vitro and in vivo (112) (312). While past efforts focused on the
development of oral protein drug delivery methods, the current study explores the
possibility of non-invasive protein drug delivery facilitated by TfR transcytosis
153
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
across the pulmonary epithelium. One of the limitations of using Tf as a carrier
molecule for pulmonary delivery of protein-based therapeutics is the predominant
localization of TfR to the basolateral membrane of alveolar epithelium (114). There
is very little transcytosis of TfR from the basolateral membrane to the apical
membrane under normal conditions. Indeed, epithelial tissues will in general have
TfR predominantly localized to the basolateral (serosal) membrane in order to
facilitate iron uptake from the blood stream. In addition, basolateral TfR is
predisposed to enter into a recycling pathway upon endocytosis, precluding the
transfer of receptors to the apical membrane. Transcytosis enhancers have
previously been utilized to overcome this dilemma to enable oral in vitro and in vivo
protein drug delivery (112; 292; 312). Transcytosis enhancers were thus considered
a means to enable Tf carrier-mediated drug delivery at the pulmonary epithelium.
BFA and T- 8 have previously been used as transcytosis enhancers. In contrast to
previously studied systems (312), T- 8 proved to rapidly destabilize the AEC
monolayer through unknown mechanisms, precluding the study of TfR-mediated
transcytosis (data not shown), while cellular tight junctions were maintained
throughout the experiment in the presence of BFA. Our efforts therefore focused on
the use of BFA as an enhancer of TfR-mediated transcytosis across alveolar
epithelium.
BFA was found not to significantly alter the polar distribution of TfR in type
n cell-like AEC monolayers (i.e., KGF-treated), as measured by binding of '^I-Tf at
4° C (Figure 27). However, BFA treatment did enhance uptake of ^®Fe-Tf from the
154
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
apical membrane and concomitantly decreased uptake of ^^Fe-Tf from the basolateral
membrane of type II cell-like AEC monolayers (Figure 27D). This apparent
discrepancy can be resolved through the following hypothesis. BFA treatment is
postulated to induce missorting of TfR, derived from the basolateral endocytic-
recycling pool or from nascently synthesized receptor, such that a portion of the TfR
is misrouted to the apical membrane where the receptor is only fleetingly situated
prior to being resorted back to the Golgi complex. In support of this hypothesis, it is
useful to note that BFA has previously been shown to reversibly alter the cis- and
trans-Golgi complexes and to disrupt normal endoplasmic reticulum-Golgi
interactions (364-366), which would tend to support the possibility that TfR could be
missorted to the apical membrane. In addition, we have previously demonstrated a
similar result when Madin-Darby canine kidney (MDCK) cell monolayers are treated
with BFA (292). The MDCK cells also exhibited similar behavior in response to
BFA treatment, with rate of uptake increased at the apical membrane without any
change in the average number of receptors present at the apical membrane (292).
The MDCK cell line used exhibits high TEER (~2500 Qcm^), characteristics similar
to the primary AEC monolayers used in the current study. Lastly, the fact that the
amount of TfR residing at the basolateral membrane was slightly diminished upon
BFA treatment (Figure 27B) in type II ceU-like monolayers, and that the level of TfR
endocytosis from the basolateral membrane was also diminished (Fig. ID), suggests
a possible receptor source-pool for the increase in TfR endocytosis from the apical
membrane. It is likely that a portion of internalized basolateral TfR would be subject
155
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
to misrouting to the apical membrane. Even though the 4° C binding studies
were unable to demonstrate a change in static apical TfR distribution in response to
BFA treatment, the ability of BFA to increase uptake of ^^Fe-Tf from the apical
surface suggested a possible means to facilitate apical-to-basolateral transcytosis in
type n cell-like AFC monolayers.
Our results indicate that BFA is able to specifically enhance apical-to-
basolateral TfR-mediated Tf transcytosis in AFC monolayers of the type II cell-like
phenotype (-f-KGF, Figure 28A), but not in AFC monolayers of the type I cell-like
phenotype (-KGF, Figure 28B). This result contrasts with previously published
reports that indicated enhancement of TfR-mediated transcytosis in AFC monolayers
presumed to be of the type I cell-like phenotype (367). AFC monolayers used in our
study had much higher TFFR values than those in the previous report (-2500 Ocm^
compared to -1300 Ocm^), suggesting a possible difference in monolayer
composition. Our results are not surprising when one considers that type II cells are
postulated to be much more metabolically active, playing a critical role in
synthesizing and secreting lung surfactant and surfactant proteins, as compared to the
relatively quiescent type I cells, whose major biological roles include formation of
the air-interface barrier and fluid clearance from the alveolar spaces, and as such
would be more likely to have a much lower iron requirement as compared to type II
cells.
Our previous results demonstrated that apical-to-basolateral transcytosis of an
insulin-Tf conjugate across Caco-2 monolayers was enhanced by BFA treatment
156
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(112). In addition, BFA enhanced the in vivo hypoglycemic effect of the insulin-Tf
conjugate when administered to streptozotocin-induced diabetic rats (312). Our
current results extend these findings to show that the TfR-mediated transcytosis
pathway is not limited to Tf conjugates covalently linked to small protein drugs like
insulin, but that it can also be utilized with much larger protein drugs like G-CSF.
Since the molecular weight of GCSF (19 kDa) is rather ubiquitous for cytokines in
general, this pathway could hold promise for many more protein drugs of interest.
We observed that a covalently linked GCSF-Tf conjugate is able to be
transcytosed across type II cell-like AEC monolayers in a manner almost identical to
that observed for Tf itself (Figure 30A). This not unexpected, since BFA-derived
enhancement of transcytosis is specific for TfR and does not influence the non
specific transport of other proteins. In support of this statement. Figure 3B
demonstrated that transport of ^^^I-GCSF in type II cell-like AEC monolayers was
not influenced by the presence of BFA. Indeed, GCSF had a substantially higher
transport across the AEC monolayer when covalently linked to Tf and subjected to
BFA exposure.
In addition to enhancement of transport, TfR-mediated transcjdosis of the
conjugate appears to deliver the protein drug to the receiver compartment in an
unaltered native state. We demonstrated that the molecular weight of the conjugate
was not altered when transcytosed via TfR-specific processes (Figure 31). A minor
fraction of degradation products were noted in the chromatographic analyses, most
likely due to the routing of a small percentage of the TfR enriched endosomes to the
157
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
lysosomal pathway. In addition, the protein drug retained biological activity when
released from the conjugate via reductive chemistry (Figure 32). Our previous
studies indicated that free insulin is released from an insulin-Tf conjugate both ex
vivo (when incubated with rat liver) and in vivo (as measured by human insulin
radioimmunoassay) (113). The GCSF-Tf conjugate utilized in this study uses
similar disulfide linker chemistry, and it is postulated that GCSF-Tf will be reduced
in a likewise fashion in vivo. In addition, the lack of substantial biological activity
for the conjugate does not necessarily obviate the potential for the monomer
conjugate to retain biological activity. The current disulfide linker chemistry results
in formation of higher-ordered cross-linked products that might potentially be
responsible for the lack of biological activity seen for the conjugate in this study.
Optimization of the cross-linking process could result in formation of conjugate
products that retain substantial biological activity. These data indicate that the
conjugate is not degraded as a result of TfR-mediated transcytosis and that the GCSF
retains biological activity. Reduction of the labile disulfide bonds within the linker
moiety liberates free active protein drug from the conjugate. This reduction
phenomenon could be predictive of in vivo conjugate reduction characteristics.
We have thus demonstrated that TfR is expressed in a polarized fashion in
monolayers of the AEC type n cell-like phenotype. In addition, it is proposed that
BFA exposure results in transient localization of TfR to the apical membrane with
subsequent rapid intemalization of these missorted receptors. This was evidenced by
the substantial increase in ^^Fe-Tf uptake at the apical surface of these monolayers in
158
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
response to BFA treatment, with the net number of receptors remaining unchanged at
the apical surface (4° C, ^^^I-Tf binding studies). BFA also enhanced TfR-mediated
transcytosis of ^^^I-Tf and the *^^I-GCSF-Tf conjugate in the apical-to-basolateral
direction across the monolayers. The conjugate was not degraded by this transport
process, and the conjugated protein retained biological activity when recovered from
the receiver compartment. This study suggests the possibility of using TfR-mediated
transcytosis for systemic delivery of therapeutic proteins via the alveolar epithelium.
Caco-2 cell culture model has come to serve as a leading qualitative indicator
in screening GI permeability and absorption of therapeutic agents (368-371). In this
report, we have investigated the amount of ^^^I-G-CSF-Tf transported in the apical-
to-basolateral direction, relative to ^^^I-G-CSF, in two-week old Caco-2 monolayers
(Figure 1). Our data demonstrate that the rate of apical-to-basolateral ^^^I-G-CSF-Tf
transport is significantly higher than that of *^^I-G-CSF (1.70 fmol/well/hr and 0.25
fmol/well/hr for ^^^I-G-CSF-Tf and ^^^I-G-CSF, respectively). The enhanced
transport of the G-CSF-Tf conjugate, compared to G-CSF, was presumed to be as a
result of TfR-mediated transcytosis processes. This was suggested by competitive
inhibition studies that demonstrated 80% reduction in ^^^I-G-CSF-Tf apical-to-
basolateral transport in the presence of 100-fold molar excess unlabeled Tf (data not
shown). We have previously demonstrated that an insulin-Tf conjugate is able to be
transcytosed across Caco-2 monolayers by TfR-specific processes, while the
unconjugated insulin was unable to be transported across Caco-2 monolayers (112).
The fact that we have demonstrated that G-CSF, a much larger protein-drug than
159
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
insulin, is also able to transported across Caco-2 monolayers as a Tf-conjugate
suggests that TfR-mediated transcytosis may serve as a multi-platform vehicle to
deliver proteins of varying sizes.
Efforts were next undertaken to examine the protein-drug downstream after
the transcytosis process. Our results indicate that G-CSF only suffered from a minor
degree of degradation as a result of the TfR-mediated transcytosis process. For
example, as shown in Figure 34, when G-CSF-Tf is recovered from the basolateral
compartment, post-TfR-mediated transport, and applied to a 40 mL S-200 column,
the major recorded peak coincides with the peak for the G-CSF-Tf standard at
fraction number 19. A low level of small molecular weight degradation products can
also be seen at fraction number 40, accounting for only 13% of the total applied
radioactivity. One can infer from this data that the molecular weight of the G-CSF-
Tf conjugate is not altered by the transcytosis process in Caco-2 monolayers and
relatively little conjugate is degraded. This result is supported by previous studies
that have shown similar results in monolayers of various cell types and for different
cargo-proteins (112; 292; 310).
In addition to determination of molecular weight, the biological activity of
the transcytosed conjugate was also determined. The conjugate was recovered after
a transcytosis experiment from the receiver compartment of Caco-2 monolayers,
concentrated ten-fold, and then subjected to a brief mild DTT reduction (25 mM
DTT, 15 min) in order to liberate free G-CSF from the conjugate. The DTT
reduction also serves to mimic the natural reduction of di-sulfide linked Tf protein-
160
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
drag conjugates that one would observe in vivo (113). As shown in Figure 35, the G-
CSF that has undergone TfR-mediated transcytosis across Caco-2 monolayers retains
almost all of its biological activity relative to control, as measured by the ability to
stimulate the proliferation of NFS-60 cells (349) (ED50 values of 0.06 ng/mL and
0.07 ng/mL for the G-CSF standard and the transcytosed-G-CSF, respectively).
Taken together, these experiments suggest that TfR-mediated transport might be able
to deliver protein-based therapeutics that are much larger than insulin (312), across
GI epithelial barriers while retaining biological function of the cargo protein.
The in vivo efficacy of G-CSF-Tf was investigated next. Acute doses of G-
CSF-Tf, filgrastim, or control vehicle were administered subcutaneously to 6 - 8 week
old BDFl mice (n=5). Absolute neutrophil counts were performed daily. As can be
seen in figure 4, the G-CSF-Tf treatment group demonstrated an increased duration
of action and significantly higher neutrophil counts (for days 2 and 3) relative to the
group receiving filgrastim. G-CSF is known to foUow a non-linear pharmacokinetic
clearance profile, which is most likely due in major part to receptor-mediated
endocytosis (372). The clearance rate of G-CSF, when covalently conjugated to Tf,
may be significantly reduced relative to filgrastim. This phenomenon could arise
due to several factors. The first is that the conjugation of G-CSF to Tf may reduce or
eliminate a predominant means of G-CSF clearance, which is dependent upon the
amount of circulating neutrophils (372). In addition, the large molecular weight of
the conjugate, relative to G-CSF, should result in reduced renal clearance, which is
the other major route of G-CSF elimination. This hypothesis is supported by
161
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
previous studies that have demonstrated a large decrease in clearance rate when G-
CSF has been genetically fused to albumin (373) or when G-CSF has been modified
by pegylation (374). In addition, the serum half-life of Tf in mice (40 h) (375) is
much longer than the terminal half-life of G-CSF (~2.5 h) (376). It is possible that
the G-CSF-Tf conjugate may have a clearance rate that is more similar to Tf than G-
CSF, as Tf is predominantly recycled after binding to its receptor. G-CSF may also
be slowly released from the conjugate as the disulfide linkage is reduced, as has been
seen for other disulfide linked protein-drug conjugates (113; 377), enabling sustained
neutrophilic leukocytosis relative to filgrastim. Another possibility is that
subcutaneous administration of G-CSF-Tf may result in the binding of the conjugate
to TfR in the interstitial tissues, creating a depot effect. G-CSF would also be slowly
released from the site of administration in this situation, resulting in a sustained
therapeutic effect.
In light of our findings that G-CSF-Tf can be actively transported across
Caco-2 monolayers (Figures 33-35), the feasibility of using G-CSF-Tf in an oral
route of administration was investigated. BDFl mice (6 - 8 week old) were given G-
CSF-Tf (50 mg/kg) or filgrastim (10 mg/kg) by gavage needle. Absolute neutrophil
counts were performed as in the subcutaneous administration studies. Oral G-CSF-
Tf was able to elicit a significant increase in neutrophil counts (for days 1 and 2)
while filgrastim had no effect when administered orally (Figure 37). In addition, G-
CSF-Tf appears to have a dose-specific response when administered orally,
providing evidence that G-CSF-Tf has a true pharmacological effect (Figure 38).
162
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Since G-CSF will only have a myelopoietic effect if it is absorbed into the
bloodstream, we presume from this data that G-CSF-Tf has a statistically significant
oral bioavailability. In addition, when comparing the area under the curve of
subcutaneously and orally administered G-CSF-Tf (Figs. 36 and 37), the oral
bioequivalence of G-CSF-Tf based only on biological activity in vivo is about 4%.
TfR is known to be highly expressed in the small intestine and Tf is absorbed by the
GI epithelium as a part of normal physiological processes (324; 378), G-CSF-Tf may
be taken up by similar processes. In addition, this result is supported by our previous
report where we demonstrated that an orally administered insulin-Tf conjugate is
able to elicit a hypoglycemic effect in diabetic rats while free insulin had no effect
(113). G-CSF-Tf may therefore be able to be orally absorbed by TfR-specific
transcellular transport processes.
We have described the creation a G-CSF-Tf conjugate that is transported
across Caco-2 monolayers by TfR-mediated transcytosis at a rate that is several-fold
higher than non-specific apical-to-basolateral G-CSF flux. G-CSF-Tf is also
recovered from the receiver compartment of Caco-2 monolayers with the molecular
weight intact and retains full biological function. The conjugate also exhibits a
prolonged myelopoietic effect in mice compared to filgrastim. This result was
observed in both subcutaneous and oral administration. The mechanism whereby G-
CSF-Tf exhibits a prolonged therapeutic effect as well as the in vivo kinetics of G-
CSF release from the conjugate remain to be determined. However, previous studies
with PEG (373) and albumin (374) have indicated that conjugation to the relatively
163
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
large molecular weight Tf may reduce clearance rate of G-CSF. In addition to the
improved pharmacokinetic characteristics of G-CSF-Tf, the development of an orally
bioavailable G-CSF has the potential to provide great benefit for patients that are
indicated for a chronic G-CSF dosing regime.
164
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
CONCLUSIONS
This report focused on the development of G-CSF-Tf conjugate as a means to
enable the transepithelial systemic delivery of G-CSF for therapeutic purposes. The
ability of the conjugate to pass through various biological barriers hinged upon the
utilization and manipulation of TfR-specific transcytosis. The transport properties of
this conjugate were investigated in in vivo systems using established cells lines (i.e.
Caco-2 and MDCK) as well as primary cultures of alveolar epithelial cells. The
effects of novel and established transcytosis enhancers on altering the polarity,
uptake, and transcytosis of TfR in these epithelial systems were also investigated. In
vivo efficacy was investigated by measuring the ability of subcutaneously and orally
delivered GCSF-Tf to stimulate the proliferation of neutrophils in BDFl mice.
The major findings of this investigation are summarized as follows:
1) G-CSF-Tf conjugate was synthesized at a 1:1 ratio using SPDP-modified Tf
coupled to DPDP-modified G-CSF. The use of DPDP enabled the free sulfhydryl
group of G-CSF’s Cysl7 to be targeted to create a defined site of linkage to Tf.
2) The G-CSF-Tf conjugate was able to be well separated from its constituent
reactant proteins using size exclusion chromatography.
165
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
3) Wortmannin, a non-specific PI-K inhibitor, treatment resulted in a significant
increase in TfR mediated transcytosis in Caco-2 monolayers. Wortmannin treatment
did not affect TEER of the Caco-2 monolayers, indicating that monolayer integrity
was maintained..
4) LY294002, a PI-K inhibitor that is specific for PD-K, treatment resulted in a
significant increase in TfR-mediated transcytosis in Caco-2 monolayers. The TEER
of the Caco-2 monolayers was not affected by LY294002 treatment.
5) When LY294002 was investigated for the ability to alter TfR distribution in
Caco-2 monolayers it was found to exhibit similar effects as the GTPase inhibitor,
AG-10, and dissimilar effects compared to BFA. For example, the amount of ^^^I-Tf
bound to the apical surface of Caco-2 monolayers at 4°C, was increased after pre
treatment with LY294002 and AG-10 (compared to non-treated monolayers), while
monolayers that received BFA pre-treatment exhibited a reduction in ^^I-Tf bound
to the apical surface.
6 ) LY294002 was found to not significantly affect TfR-mediated uptake of ^^Fe
(present in bathing fluid as ^^Fe-Tf) from the apical surface of Caco-2 monolayers
while it significantly decreased the uptake of ^^Fe-Tf from the basolateral surface of
Caco-2 monolayers. In comparison, AG-10 treatment also demonstrated no
significant alterations in TfR-mediated ^^Fe uptake from the apical surface of Caco-2
166
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
monolayers, while significantly decreasing uptake from the basolateral surface. In
contrast BFA treatment resulted in a significant increase in TfR-mediated ^^Fe
uptake from the apical surface of Caco-2 monolayers, while significantly decreasing
TfR-mediated ^^e uptake from the basolateral membrane to a greater extent than
either AG-10 or LY294002.
7) LY294002 enhanced the apical-to-basolateral transport of Tf and G-CSF-Tf
conjugate across Caco-2 monolayers more effectively than BFA and in a similar
magnitude as AG-10. The increased transport was due to an elevation of TfR
specific processes, with non-specific transport being not affected by the presence of
LY294002.
8 ) LY294002 treatment did not result in an enhancement of TfR-mediated transport
in MDCK monolayers. MDCK cells differ from Caco-2 cells in that they are thought
to only use a direct sorting mechanism from the TGN to localize TfR to the apical
membrane of the cells, while Caco-2 cells are thought to be able to also utilize an
indirect sorting mechanism that first directs TfR to the basolateral membrane before
direction to the apical surface. LY294002 was also found to have no effect on the
distribution of TfR at the apical or basolateral membrane and it did not affect the
uptake of ^^Fe-Tf from the apical or basolateral surfaces of MDCK monolayers.
LY294002 was thus theorized to affect indirect TfR sorting that is downstream of
AG-10 sensitive sorting processes.
167
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
9) The transport characteristics of G-CSF-Tf were also investigated in the
pulmonary system using primary cultured rat alveolar type n epithelial monolayers.
The primary cultured type II cell system is interesting to use because they exhibit
many in vivo like characteristics when grown on semi-permeable membranes,
including the trans-differentiation to the type I eell-like phenotype. This
investigation established that TfR is a characteristic of type II cell-like monolayers
and that TfR expression is greatly attenuated, both in cell-surface and total cell-
surface expression, after the transdifferentiation to the type I phenotype.
10) When KGF was included in the culture medium of type II monolayers, TfR
expression was maintained through culture day 8 as determined by ^^^I-Tf binding
studies and eonfocal microseopy. TfR was also expressed in a polarized fashion,
with most of the TfR being expressed at the basolateral surface and ^^Fe-Tf uptake
occurring primarily at the basolateral surface.. Since KGF has been previously
shown to maintain type II cell-like characteristics in vitro, it is assumed that these
observations are due to maintenance of type II cell-like phenotype.
11) BFA treatment was found to significantly enhance the uptake of ^®Fe-Tf from
the apical surface of rat alveolar epithelial cell monolayers. TEER was maintained
through the experiment with BFA, while AG-10 and LY294002 treatment resulted in
a rapid decrease in TEER such that monolayer barrier function was compromised.
168
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
BFA also significantly enhanced the TfR-specific transport of Tf, In-Tf, and G-CSF-
Tf across type II cell-like monolayers. BFA treatment did not affect the transport of
G-CSF alone, which was lower than G-CSF-Tf. G-CSF-Tf that was recovered from
the basolateral compartment, post-transcytosis, maintained full biological activity
after a short reduction reaction.
12) The feasibility of using G-CSF-Tf in an oral route of administration was
investigated using Caco-2 monolayers and oral administration to BDFl mice.
Conjugation of G-CSF to Tf significantly enhanced the apical-to basolateral
transport of G-CSF across Caco-2 monolayers, with biological activity of G-CSF
being retained. G-CSF-Tf administration (subcutaneous and oral) to BDFl mice
resulted in a prolonged myelopoietic effect compared to control (filgrastim).
This report has demonstrated the development of G-CSF-Tf as an efficient means to
specifically deliver G-CSF across biological barriers. The transport process is
specific to TfR-mediated transcytosis ensuring that only Tf-conjugated proteins will
pass through the biological barrier of interest. Conjugation of G-CSF to Tf may also
improve the pharmacological and pharmacokinetic properties of G-CSF. The release
of G-CSF from the conjugate complex by reduction of the disulfide linkage bonds
may result in a sustained release of G-CSF and thereby an enhanced drug half-life.
169
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
FUTURE DIRECTIONS
The future direction of this project hinges upon the ability to produce large
quantities of structurally defined product, whether with chemical or genetic linkages,
as a feasible pharmaceutical product for oral protein drug delivery. However, in the
current conjugation methodology, the Tf molecule is modified by the cross-linking
reagent, SPDP, on multiple lysine residues of variable position. It is estimated that at
least 3-5 Lys residues in Tf are equally accessible to modification by SPDP. By
using current techniques, with carefully controlled conditions, SPDP modified Tf is
usually obtained with 2-3 covalently attached SPDP groups per Tf molecule. The
end result is that Tf-protein conjugates are obtained with linkages at undetermined
lysines, creating an undefined molecular structure. In addition, even though the G-
CSF-Tf conjugate was formed with a stoichiometry of 1:1, of G-CSF to Tf, the
additional SPDP modification sites on Tf result in the formation of cross-linked
products, creating a heterogeneous mixture of variable molecular weight. This
dilemma may be solved by allowing for the formation of defined molecular species.
The success of this approach may lie in the creation of a series of fusion proteins that
will represent the next generation of Tf delivered protein drugs (figure X). The
creation of fusion proteins, consisting of defined molecular structures, will also solve
the problems presented by the current conjugation methodology, as they will be free
of the potential to form covalently linked aggregates and will be of a defined
170
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Tf
t .
GGGGGG
IgG3 hinge
G-CSF
] GGGGGG
IgG3 hinge
G-CSF
T f
Tf-G-GCSF
Tf-lgG-GCSF
GCSF-G-Tf
GGSF-lgG-Tf
Figure 42. Possible G-CSF-Tf fusion constructs. The order of the
genetic linkage is shown along with the intergenic linkage. The
smaller box within the N-terminal domain represents the respective
leader peptide signaling sequences for G-CSF and Tf.
171
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
structure. In addition, fusion protein constructs will provide a means to quickly and
efficiently produce large amounts of the orally available protein drugs. These two
themes, the creation of a defined product and rapid and efficient production, are
needed for TfR mediated delivery of protein drugs to become a reality for the
treatment of humans. The ability to non-invasively deliver these drugs will provide a
great benefit for humanity, through increased compliance to treatment regimes and
patient pain reduction.
The G-CSF-Tf fusion protein will be created by using a baculovirus
expression system. The baculovirus system has been chosen to express the fusion
protein because it provides a rapid and efficient means to produce the recombinant
protein. Nevertheless, E. coli expression is the system of choice for the production of
recombinant G-CSF because no post-translational modifications are needed for the
production of biologically active protein. However, E. coli expression for the
production of the G-CSF-Tf fusion proteins will most likely result in a functionally
inactive Tf. Bacterial expression systems for Tf have been reported, however the Tf
was biologically inactive due to improper disulfide-bond formation (379). In
contrast, the high-level production of functionally active Tf by baculovirus
expression has been previously described (380). The proteins that are produced by
the insect cells undergo post-translational processing, which is necessary for
preserving functionality in many mammalian proteins (381). In addition, the insect
cells recognize many mammalian leader signaling sequences that often greatly
increases the ease of isolation of the protein. For example, in High Five insect cells
172
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
(Life Invitrogen) the leader sequence of Tf is cleaved from the protein, to form the
mature Tf molecule, prior to the secretion into the cell culture media (382).
Therefore, it is expected that the use of baculovirus expression for the production of
G-CSF and Tf fusion constructs will result in biologically active domains for both G-
CSF andTf.
173
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
ALPHABETIZED BIBLIOGRAPHY
K. J. Addess, J. P. Basilion, R. D. Klausner, T. A. Rouault, and A. Pardi. Structure
and dynamics of the iron responsive element RNA: implications for binding of the
RNA by iron regulatory binding proteins, J. Mol. B iol, 274:72-83 (1997).
P. Aisen. Transferrin metabolism and the liver, Semin. Liver Dis., 4:193-206 (1984).
P. Aisen and E. B. Brown. Structure and function of transferrin. Prog. Hematol,
9:25-56 (1975).
P. Aisen and E. B. Brown. The iron-binding function of transferrin in iron
metabolism, Semin. Hematol, 14:31-53 (1977).
P. Aisen, A. Leibman, and R. A. Pinkowitz. The anion-binding functions of
transferrin, Adv. Exp. Med. B iol, 48:125-140 (1974).
P. Aisen and I. Listowsky. Iron transport and storage proteins, Annu. Rev. Biochem.,
49:357-393 (1980).
C. E. Alford, T. E. King, Jr., and P. A. Campbell. Role of transferrin, transferrin
receptors, and iron in macrophage listericidal activity, J. Exp. Med., 174:459-466
(1991).
Alper, C. A., Raum D., Awdeh Z.L., Petersen B.H., Taylor P.D., and Starzl T.E.
Studies of hepatic synthesis in vivo of plasma proteins, including orosomucoid,
transferrin, antitrypsin, C8 , and factor Bl. Clin.Immunol.Immunopathol. 16, 84-89
(1974).
D. R. Ambruso and R. B. Johnston, Jr. Lactoferrin enhances hydroxyl radical
production by human neutrophils, neutrophil particulate fractions, and an enzymatic
generating system, J. Clin. Invest, 67:352-360 (1981).
174
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
N. C. Andrews. Disorders of iron metabolism, N. Engl. J. Med., 341:1986-1995
(1999).
G. Apodaca, B. Aroeti, K. Tang, and K. Mostov. Brefeldin-A inhibits the delivery of
the polymeric immunoglobulin receptor to the basolateral surface of MDCK cells., J.
Biol. Chem., 268:20380-20385 (1993).
K. L. Audus, R. L. Bartel, I. J. Hidalgo, and R. T. Borchart. The use of cultured
epithelial and endothelial cells for drug transport and metabolism studies., Pharm.
Res., 7:435-451 (1990).
Azari, K. L. and Feeney, R. E. Resistance of metal complexes of conalbumin and
transferrin to proteolysis and thermal denaturation. J.Biol.Chem. 232,293-302. 1958.
Ref Type: Generic
Baggiolilni, A., Deduve, C., Masson, P. L., and Heremans, J. F. Association of
lactoferrin with specific granules in rabbit heterophil leucocytes. J.Exp.Med. 131,
559-570 (1970).
E. N. Baker and P. F. Lindley. New perspectives on the structure and function of
transferrins, J. Inorg. Biochem., 47:147-160 (1992).
D. Banerjee, P. R. Flanagan, J. Cluett, and L. S. Valberg. Transferrin receptors in the
human gastrointestinal tract., Gastroenterol, 91:861-869 (1986).
D. Baneqee, P. R. Flanagan, J. Cluett, and L. S. Valberg. Transferrin receptors in the
human gastrointestinal tract. Relationship to body iron stores. Gastroenterology,
91:861-869 (1986).
J. P. Basilion, M. C. Kennedy, H. Beinert, C. M. Massinople, R. D. Klausner, and T.
A. Rouault. Overexpression of iron-responsive element-binding protein and its
analytical characterization as the RNA-binding form, devoid of an iron- sulfur
cluster. Arch. Biochem. Biophys., 311:517-522 (1994).
1 7 5
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
J. M. Bastin, M. Jones, C. A. O'Callaghan, L. Schimanski, D. Y. Mason, and A. R.
Townsend. Kupffer cell staining by an HFE-specific monoclonal antibody:
implications for hereditary haemochromatosis, Br. J. Haematol, 103:931-941
(1998).
G. W. Bates and M. R. Schlabach. The nonspecific binding of Fe3+ to transferrin in
the absence of synergistic anions, J. Biol Chem., 250:2177-2181 (1975).
Y. Beguin, S. Lampertz, D. De Groote, D. Igot, M. Malaise, and G. Fillet. Soluble
CD23 and other receptors (CD4, CD8, CD25, CD71) in serum of patients with
chronic lymphocytic leukemia. Leukemia, 7:2019-2025 (1993).
N. Bejaoui, M. Page, and C. Noel. Cytotoxicity of transferrin-daunorubicin
conjugates on small cell carcinoma of the lung (SCCL) cell line NCI-H69,
Anticancer Res., 11:2211-2213 (1991).
D. Y. Bell, J. A. Haseman, A. Spock, G. McLennan, and G. E. Hook. Plasma
proteins of the bronchoalveolar surface of the lungs of smokers and nonsmokers. Am.
Rev. Respir. Dis., 124:72-79 (1981).
M. J. Bennett, J. A. Lebron, and P. J. Bjorkman. Crystal structure of the hereditary
haemochromatosis protein HFE complexed with transferrin receptor. Nature,
403:46-53 (2000).
L. S. Bernstein, A. A. Grillo, S. S. Loranger, and M. E. Linder. RGS4 binds to
membranes through an amphipathic alpha -helix, J. Biol Chem., 275:18520-18526
(2000).
C. R. Bhasker, G. Burgiel, B. Neupert, A. Emery-Goodman, L. C. Kuhn, and B. K.
May. The putative iron-responsive element in the human erythroid 5-
aminolevulinate synthase mRNA mediates translational control, J. Biol Chem.,
268:12699-12705 (1993).
176
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
U. Bickel, T. Yoshikawa, E. M. Landaw, K. F. Faull, and W. M. Pardridge.
Pharmacologic effects in vivo in brain by vector-mediated peptide drug delivery,
Proc. Natl. Acad. Sci. U. S. A, 90:2618-2622 (1993).
U. Bickel, T. Yoshikawa, and W. M. Pardridge. Delivery of peptides and proteins
through the blood-brain barrier, Adv. Drug Deliv. Rev., 46:247-279 (2001).
H. Bim, P. J. Verroust, E. Nexo, H. Hager, C. Jacobsen, E. I. Christensen, and S. K.
Moestrup. Characterization of an epithelial approximately 460-kDa protein that
facilitates endocytosis of intrinsic factor-vitamin B12 and binds receptor-associated
protein, /. Biol. Chem., 272:26497-26504 (1997).
B. Bloch, T. Popovici, S. Chouham, M. J. Levin, D. Tuil, and A. Kahn. Transferrin
gene expression in choroid plexus of the adult rat brain. Brain Res. Bull., 18:573-576
(1987).
B. Bloch, T. Popovici, M. J. Levin, D. Tuil, and A. Kahn. Transferrin gene
expression visualized in oligodendrocytes of the rat brain by using in situ
hybridization and immunohistochemistry, Proc. Natl. Acad. Sci. U. S. A, 82:6706-
6710(1985).
P. Bork and G. Beckmann. The CUB domain. A widespread module in
developmentally regulated proteins, J. Mol. Biol., 231:539-545 (1993).
Z. Borok, S. I. Danto, R. L. Lubman, Y. Cao, M. C. Williams, and E. D. Crandall.
Modulation of tl alpha expression with alveolar epithelial cell phenotype in vitro.
Am. J. Physiol, 275:L155-L164 (1998).
Z. Borok, S. I. Danto, S. M. Zabski, and E. D. Crandall. Defined medium for primary
culture de novo of adult rat alveolar epithelial cells. In Vitro Cell Dev. Biol. Anim,
30A:99-104 (1994).
Z. Borok, J. M. Liebler, R. L. Lubman, M. J. Foster, B. Zhou, X. Li, S. M. Zabski, K.
J. Kim, and E. D. Crandall. Na transport proteins are expressed by rat alveolar
epithelial type I cells. Am. J. Physiol Lung Cell Mol. Physiol, 282:L599-L608
(2002).
177
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Z. Borok, R. L. Lubman, S. I. Danto, X. L. Zhang, S. M. Zabski, L. S. King, D. M.
Lee, P. Agre, and E. D. Crandall. Keratinocyte growth factor modulates alveolar
epithelial cell phenotype in vitro: expression of aquaporin 5, Am. J. Respir. Cell Mol.
Biol., 18:554-561 (1998).
G. Bottger, B. Nagelkerken, and S. P. van der. Rab4 and Rab7 define distinct
nonoverlapping endosomal compartments, J. Biol. Chem., 271:29191-29197 (1996).
C. Bouton, M. J. Chauveau, S. Lazereg, and J. C. Drapier. Recycling of RNA
binding iron regulatory protein 1 into an aconitase after nitric oxide removal depends
on mitochondrial ATP, J. Biol. Chem., 277:31220-31227 (2002).
P. P. Breitfeld, J. E. Casanova, N. E. Simister, S. A. Ross, W. C. McKinnon, and K.
E. Mostov. Sorting signals, Curr. Opin. Cell Biol, 1:617-623 (1989).
N. A. Bright, M. R. Lindsay, A. Stewart, and J. P. Luzio. The relationship between
lumenal and limiting membranes in swollen late endocjhic compartments formed
after wortmannin treatment or sucrose accumulation, Traffic.,631-642 (2001).
B. E. Britigan, M. B. Hayek, B. N. Doebbeling, and R. B. Pick, Jr. Transferrin and
lactoferrin undergo proteolytic cleavage in the Pseudomonas aeruginosa-infected
lungs of patients with cystic fibrosis, Infect. Immun., 61:5049-5055 (1993).
J. H. Brock and I. Esparza. Failure of reticulocytes to take up iron from lactoferrin
saturated by various methods, Br. J. Haematol, 42:481-483 (1979).
I. P. Brown, R. M. Hewick, I. Hellstrom, K. E. Hellstrom, R. F. Doolittle, and W. J.
Dreyer. Human melanoma-associated antigen p97 is stracturally and functionally
related to transferrin. Nature, 296:171-173 (1982).
W. J. Brown, D. B. DeWald, S. D. Emr, H. Plutner, and W. E. Balch. Role for
phosphatidylinositol 3-kinase in the sorting and transport of newly synthesized
lysosomal enzymes in mammalian cells, J. Cell Biol, 130:781-796 (1995).
C. Bucci, A. Lutcke, O. Steele-Mortimer, V. M. Olkkonen, P. Dupree, M. Chiariello,
C. B. Bruni, K. Simons, and M. Zerial. Co-operative regulation of endocytosis by
three Rab5 isoforms, FEBSLett., 366:65-71 (1995).
178
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
B. Buendia, M. H. Bre, G. Griffiths, and E. Karsenti. Cytoskeletal control of
centrioles movement during the establishment of polarity in Madin-Darby canine
kidney cells, J. Cell Biol, 110:1123-1135 (1990).
J. J. Bullen and J. A. Armstrong. The role of lactoferrin in the bactericidal function
of polymorphonuclear leucocytes. Immunology, 36:781-791 (1979).
C. G. Burd and S. D. Emr. Phosphatidylinositol(3)-phosphate signaling mediated by
specific binding to RING FYVE domains, M ol Cell, 2:157-162 (1998).
C. Camaschella, A. Roetto, A. Cali, M. De Gobbi, G. Garozzo, M. Carella, N.
Majorano, A. Totaro, and P. Gasparini. The gene TFR2 is mutated in a new type of
haemochromatosis mapping to 7q22, Nat. Genet., 25:14-15 (2000).
C. Candiani, M. Tommasi, G. Fracasso, I. Lorenzetti, A. Adami, G. Benoni, G.
Tridente, and M. Colombatti. Pharmacokinetics of intrathecal transferrin-ricin a
chain immunotoxin. Life Sci, 69:335-346 (2001).
J. E. Casanova, X. Wang, R. Kumar, S. G. Bhartur, J. Navarre, J. E. Woodrum, Y.
Altschuler, G. S. Ray, and J. R. Goldenring. Association of Rab25 and R ablla with
the apical recycling system of polarized Madin-Darby canine kidney cells, Mol Biol
Cell,41-6l (1999).
J. L. Casey, M. W. Hentze, D. M. Koeller, S. W. Caughman, T. A. Rouault, R. D.
Klausner, and J. B. Harford. Iron-responsive elements: regulatory RNA sequences
that control mRNA levels and translation. Science, 240:924-928 (1988).
B. P. Ceresa, M. Lotscher, and S. L. Schmid. Receptor and membrane recycling can
occur with unaltered efficiency despite dramatic Rab5(q791)-induced changes in
endosome geometry, J. Biol Chem., 276:9649-9654 (2001).
P. A. Charlwood. Differential sedimentation-velocity and gel-flltration
measurements on human apotransferrin and iron-transferrin, Biochem. J., 125:1019-
1026(1971).
179
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
F. Chatelet, E. Brianti, P. Ronco, J. Roland, and P. Verroust. Ultrastructural
localization by monoclonal antibodies of brush border antigens expressed by
glomeruli. I. Renal distribution. Am. J. Pathol, 122:500-511 (1986).
F. Chatelet, E. Brianti, P. Ronco, J. Roland, and P. Verroust. Ultrastmctural
localization by monoclonal antibodies of brush border antigens expressed by
glomeruli, n. Extrarenal distribution. Am. J. Pathol, 122:512-519 (1986).
P. Chavrier, S. P. van der, Z. Mishal, B. Nagelkerken, and J. P. Gorvel. Early
endosome membrane dynamics characterized by flow cytometry. Cytometry, 29:41-
49 (1997).
J. M. Cheek, M. J. Evans, and E. D. Crandall. Type I cell-like morphology in tight
alveolar epithelial monolayers. Exp. Cell Res., 184:375-387 (1989).
C. Chen, K. T. Seow, K. Guo, L. P. Yaw, and S. C. Lin. The membrane association
domain of RGS16 contains unique amphipathic features that are conserved in RGS4
and RGS5, J. Biol Chem., 274:19799-19806 (1999).
W. Chen, Y. Feng, D. Chen, and A. Wandinger-Ness. Rabl 1 is required for trans-
golgi network-to-plasma membrane transport and a preferential target for GDP
dissociation inhibitor. Mol Biol Cell, 9:3241-3257 (1998).
W. J. Chen, J. L. Goldstein, and M. S. Brown. NPXY, a sequence often found in
cytoplasmic tails, is required for coated pit-mediated internalization of the low
density lipoprotein receptor, J. Biol Chem. , 265:3116-3123 (1990).
P. W. Cheng. Receptor ligand-facilitated gene transfer: enhancement of liposome-
mediated gene transfer and expression by transferrin. Hum. Gene Ther., 7:275-282
(1996).
M. Chiariello, C. B. Bruni, and C. Bucci. The small GTPases Rab5a, Rab5b and
Rab5c are differentially phosphorylated in vitro, F E B S Lett., 453:20-24 (1999).
E. I. Christensen, J. Gliemann, and S. K. Moestrup. Renal tubule gp330 is a calcium
binding receptor for endocytic uptake of protein, J. Histochem. Cytochem., 40:1481-
1490 (1992).
180
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
s. Christoforidis, H. M. McBride, R. D. Burgoyne, and M. Zerial. The Rab5 effector
EEAl is a core component of endosome docking, Nature, 397:621-625 (1999).
S. Christoforidis, M. Miaczynska, K. Ashman, M. Wilm, L. Zhao, S. C. Yip, M. D.
Waterfield, J. M. Backer, and M. Zerial. Phosphatidylinositol-3-OH kinases are
Rab5 effectors, Nat. Cell Biol, 1:249-252 (1999).
L. Chores, J. M. Ferreras, R. Munoz, J. Benitez, P. Jimenez, and T. Girbes. Targeting
cancer cells with transferrin conjugates containing the non- toxic type 2 ribosome-
inactivating proteins nigrin h or ebulin 1 , Cancer Lett., 184:29-35 (2002).
A. Constable, S. Quick, N. K. Gray, and M. W. Hentze. Modulation of the RNA-
binding activity of a regulatory protein by iron in vitro: switching between enzymatic
and genetic function?, Proc. N atl Acad. Set U. S. A, 89:4554-4558 (1992).
B. Corsi, S. Levi, A. Cozzi, A. Corti, D. Altimare, A. Albertini, and P. Arosio.
Overexpression of the hereditary hemochromatosis protein, HFE, in HeLa cells
induces and iron-deficient phenotype, FEBS Lett., 460:149-152 (1999).
M. Gotten, F. Langle-Rouault, H. Kirlappos, E. Wagner, K. Mechtler, M. Zenke, H.
Beug, and M. L. Bimstiel. Transferrin-polycation-mediated introduction of DNA
into human leukemic cells: stimulation by agents that affect the survival of
transfected DNA or modulate transferrin receptor levels, Proc. N atl Acad. Sci U. S.
A, 87:4033-4037 (1990).
R. R. Crichton. Proteins of iron storage and transport. Adv. Protein Chem., 40:281-
363 (1990).
H. Damke, J. Klumperman, K. von Figura, and T. Braulke. Brefeldin A affects the
cellular distribution of endocytic receptors differentially, Biochem. Biophys. Res.
Common., 185 :719-727 (1992).
S. I. Danto, J. M. Shannon, Z. Borok, S. M. Zabski, and E. D. Crandall. Reversible
transdifferentiation of alveolar epithelial cells. Am. J. Respir. Cell M ol Biol,
12:497-502 (1995).
181
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
E. Daro, S. P. van der, T. Galli, and I. Mellman. Rab4 and cellubrevin define
different early endosome populations on the pathway of transferrin receptor
recycling, Proc. Natl. Acad. Sci. U. S. A,9559-9564 (1996).
P. R. Dash, M. L. Read, K. D. Fisher, K. A. Howard, M. Wolfert, D. Oupicky, V.
Subr, J. Strohalm, K. Ulbrich, and L. W. Seymour. Decreased binding to proteins
and cells of polymeric gene delivery vectors surface modified with a multivalent
hydrophilic polymer and retargeting through attachment of transferrin, J. Biol.
Chem., 275:3793-3802 (2000).
A. Dautry-Varsat. Receptor-mediated endocytosis: the intracellular joumey of
transferrin and its receptor., Biochimie, 68:375-381 (1986).
M. Davies, J. E. Parry, and R. G. Sutcliffe. Examination of different preparations of
human placental plasma membrane for the binding of insulin, transferrin and
immunoglobulins, J. Reprod. Fertil, 63:315-324 (1981).
C. G. Davis, J. L. Goldstein, T. C. Sudhof, R. G. Anderson, D. W. Russell, and M. S.
Brown. Acid-dependent ligand dissociation and recycling of LDL receptor mediated
by growth factor homology region. Nature, 326:760-765 (1987).
R. J. Davis, S. Corvera, and M. P. Czech. Insulin stimulates cellular iron uptake and
causes the redistribution of intracellular transferrin receptors to the plasma
membrane, J. Biol. Chem., 261:8708-8711 (1986).
M. De Gobbi, F. Daraio, C. Oberkanins, A. Moritz, F. Kury, G. Fiorelli, and C.
Camaschella. Analysis of HFE and TFR2 mutations in selected blood donors with
biochemical parameters of iron overload, Haematologica, 88:396-401 (2003).
M. C. de Lima, S. Simoes, P. Pires, R. Caspar, V. Slepushkin, and N. Duzgunes.
Gene delivery mediated by cationic liposomes: from biophysical aspects to
enhancement of transfection. Mol. Membr. Biol, 16:103-109 (1999).
C. J. Dekker, M. J. Kroos, H. C. Van der, and H. G. Van Eijk. Uptake of sialo and
asialo transferrins by isolated rat hepatocytes. Comparison of a heterologous and a
homologous system. Int. J. Biochem., 17:701-706 (1985).
182
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
p. A. DeRusso, C. C. Philpott, K. Iwai, H. S. Mostowski, R. D. Klausner, and T. A.
Rouault. Expression of a constitutive mutant of iron regulatory protein 1 abolishes
iron homeostasis in mammalian cells, J. Biol. Chem., 270:15451-15454 (1995).
D. Deshpande, D. Toledo-Velasquez, L. Y. Wang, C. J. Malanga, J. K. Ma, and Y.
Rojanasakul. Receptor-mediated peptide delivery in pulmonary epithelial
monolayers, Pharm. Res., 1 1 :1121-1126 (1994).
L. G. Dobbs, M. C. Williams, and R. Gonzalez. Monoclonal antibodies specific to
apical surfaces of rat alveolar type I cells bind to surfaces of cultured, but not freshly
isolated, type II cells, Biochim. Biophys. Acta, 970:146-156 (1988).
R. Doms, G. Russ, and J. Yewdell. Brefeldin A redistributes resident and itinerant
Golgi proteins to endoplasmic reticulum., J. Cell Biol, 109:61-72 (1989).
V. Dubljevic, A. Sali, and J. W. Goding. A conserved RGD (Arg-Gly-Asp) motif in
the transferrin receptor is required for binding to transferrin, Biochem. J., 341 ( Ft
1): 11-14 (1999).
A. Durrbach, D. Louvard, and E. Coudrier. Actin filaments facilitate two steps of
endocytosis, J. Cell Set, 109 ( Ft 2):457-465 (1996).
A. Durrbach, G. Raposo, D. Tenza, D. Louvard, and E. Coudrier. Truncated brush
border myosin I affects membrane traffic in polarized epithelial cells. Traffic.,411-
424 (2000).
A. Egyed. The significance of transferrin-bound bicarbonate in the uptake of iron by
reticulocytes, Biochim. Biophys. Acta, 304:805-813 (1973).
C. A. Enns, J. E. Shindelman, S. E. Tonik, and H. H. Sussman.
Radioimmunochemical measurement of the transferrin receptor in human trophoblast
and reticulocyte membranes with a specific anti-receptor antibody, Proc. Natl Acad.
Sci U. S. A, 78:4222-4225 (1981).
C. A. Enns and H. H. Sussman. Physical characterization of the transferrin receptor
in human placentae, J. Biol Chem., 256:9820-9823 (1981).
183
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
C. A. Enns and H. H. Sussman. Similarities between the transferrin receptor proteins
on human reticulocytes and human placentae, J. Biol. Chem., 256:12620-12623
(1981).
R. Erlitzki, J. C. Long, and E. C. Theil. Multiple, conserved iron-responsive elements
in the 3'-untranslated region of transferrin receptor mRNA enhance binding of iron
regulatory protein 2, J. Biol. Chem., 277:42579-42587 (2002).
W. P. Faulk, B. L. Hsi, and P. J. Stevens. Transferrin and transferrin receptors in
carcinoma of the breast, Lancet, 2:390-392 (1980).
J. N. Feder, A. Gnirke, W. Thomas, Z. Tsuchihashi, D. A. Ruddy, A. Basava, F.
Dormishian, R. Domingo, Jr., M. C. Ellis, A. Fullan, L. M. Hinton, N. L. Jones, B. E.
Kimmel, G. S. Kronmal, P. Lauer, V. K. Lee, D. B. Loeb, F. A. Mapa, E.
McClelland, N. C. Meyer, G. A. Mintier, N. Moeller, T. Moore, E. Morikang, R. K.
Wolff, and . A novel MHC class I-like gene is mutated in patients with hereditary
haemochromatosis, Nat. Genet., 13:399-408 (1996).
J. N. Feder, D. M. Penny, A. hxinki, V. K. Lee, J. A. Lebron, N. Watson, Z.
Tsuchihashi, E. Sigal, P. J. Bjorkman, and R. C. Schatzman. The hemochromatosis
gene product complexes with the transferrin receptor and lowers its affinity for
ligand binding, Proc. Natl. Acad. Sci. U.S. A, 95:1472-1477 (1998).
J. N. Feder, Z. Tsuchihashi, A. Irrinki, V. K. Lee, F. A. Mapa, E. Morikang, C. E.
Prass, S. M. Starnes, R. K. Wolff, S. Parkkila, W. S. Sly, and R. C. Schatzman. The
hemochromatosis founder mutation in HLA-H disrupts beta2- microglobulin
interaction and cell surface expression, J. Biol. Chem., 272:14025-14028 (1997).
K. D. Fisher, K. Ulbrich, V. Subr, C. M. Ward, V. Mautner, D. Blakey, and L. W.
Seymour. A versatile system for receptor-mediated gene delivery permits increased
entry of DNA into target cells, enhanced delivery to the nucleus and elevated rates of
transgene expression. Gene Ther., 7:1337-1343 (2000).
R. E. Fleming, M. C. Migas, C. C. Holden, A. Waheed, R. S. Britton, S. Tomatsu, B.
R. Bacon, and W. S. Sly. Transferrin receptor 2: continued expression in mouse liver
in the face of iron overload and in hereditary hemochromatosis, Proc. Natl. Acad.
Sci. U. S. A, 97:2214-2219 (2000).
184
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
R. E. Fleming and W. S. Sly. Hepcidin; a putative iron-regulatory hormone relevant
to hereditary hemochromatosis and the anemia of chronic disease., Proc. Natl Acad.
Sci. USA, 98:8160-8162 (2001).
M. R. Food, E. O. Sekyere, and D. R. Richardson. The soluble form of the
membrane-bound transferrin homologue, melanotransferrin, inefficiently donates
iron to cells via nonspecific internalization and degradation of the protein, Eur. ./.
Biochem., 269:4435-4445 (2002).
P. M. Friden. Receptor-mediated transport of therapeutics across the blood-brain
barrier, Neurosurgery, 35:294-298 (1994).
M. Fritzer, T. Szekeres, V. Szuts, H. N. Jarayam, and H. Goldenberg. Cytotoxic
effects of a doxorubicin-transferrin conjugate in multidrag- resistant KB cells,
Biochem. Pharmacol, 51:489-493 (1996).
J. Fuchs, M. Fodda, L. Packer, and R. Kaufmann. Morbidity risk in HFE associated
hereditary hemochromatosis C282Y heterozygotes, Toxicology, 180:169-181 (2002).
T. Fujiwara, K. Oda, S. Yokota, A. Takatsuki, and Y. Ikehara. Brefeldin A causes
disassembly of the Golgi complex and accumulation of secretory proteins in the
endoplasmic reticulum, J. Biol Chem., 263:18545-18552 (1988).
C. E. Futter, A. Gibson, E. H. Allchin, S. Maxwell, L. J. Ruddock, G. Odorizzi, D.
Domingo, I. S. Trowbridge, and C. R. Hopkins. In polarized MOCK cells basolateral
vesicles arise from clathrin-gamma- adaptin-coated domains on endosomal tubules,
J. Cell B iol, 141:611-623 (1998).
R. Gagescu, N. Demaurex, R. G. Parton, W. Hunziker, L. A. Huber, and J.
Graenberg. The recycling endosome of Madin-Darby canine kidney cells is a mildly
acidic compartment rich in raft components. Mol. Biol Cell, 11:2775-2791 (2000).
L. L. Gan and D. R. Thakker. Application of the Caco-2 model in the design and
development of orally active drags: elucidation of biochemical and physiological
barriers posed by the intestinal epithium.. Adv. Drug Delivery Rev., 23:77-98 (1997).
185
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
K. C. Gatter, G. Brown, I. S. Trowbridge, R. E. Woolston, and D, Y. Mason.
Transferrin receptors in human tissues: their distribution and possible clinical
relevance, J. Clin. Pathol, 36:539-545 (1983).
J. M. Gaullier, A. Simonsen, A. D'Anigo, B. Bremnes, H. Stenmark, and R. Aasland.
FYVE fingers bind PtdIns(3)P, Nature, 394:432-433 (1998).
J. Gautron, M. T. Hincke, M. Panheleux, J. M. Garcia-Ruiz, T. Boldicke, and Y.
Nys. Ovotransferrin is a matrix protein of the hen eggshell membranes and basal
calcified layer. Connect. Tissue Res., 42:255-267 (2001).
F. Giansanti, P. Rossi, M. T. Massucci, D. Botti, G. Antonini, P. Valenti, and L.
Seganti. Antiviral activity of ovotransferrin discloses an evolutionary strategy for the
defensive activities of lactoferrin, Biochem. Cell B iol, 80:125-130 (2002).
D. J. Gillooly, A. Simonsen, and H. Stenmark. Cellular functions of
phosphatidylinositol 3-phosphate and FYVE domain proteins, Biochem. J., 355:249-
258 (2001).
J. W. Goding and G. F. Burns. Monoclonal antibody OKT-9 recognizes the receptor
for transferrin on human acute lymphocytic leukemia cells, J. Immunol, 127:1256-
1258 (1981).
J. R. Goldenring, J. Smith, H. D. Vaughan, P. Cameron, W. Hawkins, and J.
Navarre. R abll is an apically located small GTP-binding protein in epithelial tissues.
Am J Physiol, 270 :G515-G525 (1996).
I. Graham and J. Williams. A comparsion of glycopeptides from the transferrins of
several species, Biochem. J., 145:263-279 (1975).
E. G. Green, E. Ramm, N. M. Riley, D. J. Spiro, J. R. Goldenring, and M. Wessling-
Resnick. Rabl 1 is associated with transferrin-containing recycling compartments in
K562 cells, Biochem. Biophys. Res. Commun., 239:612-616 (1997).
1 8 6
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
C. N. Gross, A. Irrinki, J. N. Feder, and C. A. Enns. Co-trafficking of HFE, a
nonclassical major histocompatibility complex class I protein, with the transferrin
receptor implies a role in intracellular iron regulation, J. Biol. Chem., 273:22068-
22074 (1998).
J. G. Grossmann, A. B. Mason, R. C. Woodworth, M. Neu, P. F. Lindley, and S. S.
Hasnain. Asp ligand provides the trigger for closure of transferrin molecules. Direct
evidence from X-ray scattering studies of site-specific mutants of the N-terminal
half-molecule of human transferrin, J. Mol. B iol, 231:554-558 (1993).
B. Guo, F. M. Brown, J. D. Phillips, Y. Yu, and E. A. Leibold. Characterization and
expression of iron regulatory protein 2 (IRP2). Presence of multiple IRP2 transcripts
regulated by intracellular iron levels, J. Biol Chem., 270:16529-16535 (1995).
B. Guo, J. D. Phillips, Y. Yu, and E. A. Leibold. Iron regulates the intracellular
degradation of iron regulatory protein 2 by the proteasome, J. Biol Chem.,
270:21645-21651 (1995).
B. Guo, Y. Yu, and E. A. Leibold. Iron regulates cytoplasmic levels of a novel iron-
responsive element- binding protein without aconitase activity, J. Biol Chem.,
269:24252-24260 (1994).
M. Guo, H. Sun, H. J. McArdle, L. Gambling, and P. J. Sadler. Ti(IV) uptake and
release by human serum transferrin and recognition of Ti(IV)-transferrin by cancer
cells: understanding the mechanism of action of the anticancer drug titanocene
dichloride. Biochemistry, 39:10023-10033 (2000).
J. M. Gutteridge, S. Mumby, G. J. Quinlan, K. F. Chung, and T. W. Evans. Pro
oxidant iron is present in human pulmonary epithelial lining fluid: implications for
oxidative stress in the lung, Biochem. Biophys. Res. Commun., 220:1024-1027
(1996).
N. Hagihara, S. Walbridge, A. W. Olson, E. H. Oldfield, and R. J. Youle. Vascular
protection by chloroquine during brain tumor therapy with Tf- CRM107, Cancer
Res., 60:230-234 (2000).
187
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
D. J. Haile, T. A. Rouault, C. K. Tang, J. Chin, J. B. Harford, and R. D. Klausner.
Reciprocal control of RNA-binding and aconitase activity in the regulation of the
iron-responsive element binding protein: role of the iron-sulfur cluster, Proc. Natl.
Acad. Sci. U. S. A, 89:7536-7540 (1992).
C. M. Hales, R. Griner, K. C. Hobdy-Henderson, M. C. Dorn, D. Hardy, R. Kumar,
J. Navarre, E. K. Chan, L. A. Lapierre, and J. R. Goldenring. Identification and
characterization of a family of Rab 11-interacting proteins, J. Biol. Chem.,
276:39067-39075 (2001).
W. Halpem, T. A. Riccobene, H. Agostini, K. Baker, D. Stolow, M. L. Gu, J. Hirsch,
A. Mahoney, J. Carrell, E. Boyd, and K. J. Grzegorzewski. Albugranin, a
recombinant human granulocyte colony stimulating factor (G-CSF) genetically fused
to recombinant human albumin induces prolonged myelopoietic effects in mice and
monkeys, Pharm. Res., 19:1720-1729 (2002).
T. A. Hamilton, H. G. Wada, and H. H. Sussman. Identification of transferrin
receptors on the surface of human cultured cells, Proc. Natl. Acad. Sci. U. S. A,
76:6406-6410 (1979).
J. Harford, K. Bridges, G. Ashwell, and R. D. Klausner. Intracellular dissociation of
receptor-bound asialoglycoproteins in cultured hepatocytes. A pH-mediated
nonlysosomal event, J. Biol. Chem., 258:3191-3197 (1983).
Q. Y. He, A. B. Mason, B. M. Tam, R. T. MacGillivray, and R. C. Woodworth. Dual
role of Lys206-Lys296 interaction in human transferrin N-lobe: iron-release trigger
and anion-binding site. Biochemistry, 38:9704-9711 (1999).
F. Heckner. Practical Microscopic Hematology , Urban & Schwarzenberg,
Baltimore, 1982.
B. R. Henderson and L. C. Kuhn. Differential modulation of the RNA-binding
proteins IRP-1 and IRP-2 in response to iron. IRP-2 inactivation requires translation
of another protein, J. Biol. Chem., 270:20509-20515 (1995).
188
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
B. R. Henderson, C. Seiser, and L. C. Kuhn. Characterization of a second RNA-
binding protein in rodents with specificity for iron-responsive elements, J. Biol.
Chem., 268:27327-27334 (1993).
M. W. Hentze, S. W. Caughman, T. A. Rouault, J. G. Barriocanal, A. Dancis, J. B.
Harford, and R. D. Klausner. Identification of the iron-responsive element for the
translational regulation of human ferritin mRNA, Science, 238:1570-1573 (1987).
M. W. Hentze, T. A. Rouault, S. W. Caughman, A. Dancis, J. B. Harford, and R. D.
Klausner. A cis-acting element is necessary and sufficient for translational regulation
of human ferritin expression in response to iron, Proc. Natl. Acad. Sci. U. S. A,
84:6730-6734 (1987).
I. J. Hidalgo, T. J. Raub, and R. T. Borchardt. Characterization of the human colon
carcinoma cell line (Caco-2) as a model system for intestinal epitheMal permeability,
Gastroenterology, 96:736-749 (1989).
G. Hjalm, E. Murray, G. Crumley, W. Harazim, S. Lundgren, I. Onyango, B. Ek, M.
Larsson, C. Juhlin, P. Heilman, H. Davis, G. Akerstrom, L. Rask, and B. Morse.
Cloning and sequencing of human gp330, a Ca(2+)-binding receptor with potential
intracellular signaling properties, Eur. J. Biochem., 239:132-137 (1996).
J. F. Holter, J. E. Weiland, E. R. Pacht, J. E. Gadek, and W. B. Davis. Protein
permeability in the adult respiratory distress syndrome. Loss of size selectivity of the
alveolar epithelium, J. Clin. Invest, 78:1513-1522 (1986).
H. Huebers, E. Huebers, W. Rummel, and R. R. Crichton. Isolation and
characterization of iron-binding proteins from rat intestinal mucosa, Eur. J.
Biochem., 66:447-455 (1976).
H. A. Huebers, E. Csiba, E. Huebers, and C. A. Finch. Competitive advantage of
diferric transferrin in delivering iron to reticulocytes, Proc. Natl. Acad. Sci. U. S. A,
80:300-304 (1983).
H. A. Huebers and C. A. Finch. The physiology of transferrin and transferrin
receptors. Physiol Rev., 67:520-582 (1987).
189
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
H. A. Huebers and C. A. Finch. The physiology of transferrin and transferrin
receptors. Physiol Rev., 67:520-582 (1987).
L. Hunyady, A. J. Baukal, Z. Gaborik, J. A. Olivares-Reyes, M. Bor, M. Szaszak, R.
Lodge, K. J. Catt, and T. Balia. Differential PI 3-kinase dependence of early and late
phases of recycling of the internalized ATI angiotensin receptor, J. Cell Biol,
157:1211-1222 (2002).
W. Hunziker, J. A. Whitney, and I. Mellman. Selective inhibition of transcytosis by
brefedin A in MDCK cells.. Cell, 67:617-627 (1991).
J. Huwyler, A. Cerletti, G. Pricker, A. N. Eberle, and J. Drewe. By-passing of P-
glycoprotein using immunoliposomes, J. Drug Target, 10:73-79 (2002).
J. Huwyler, D. Wu, and W. M. Pardridge. Brain drug delivery of small molecules
using immunoliposomes, Proc. N atl Acad. Sci. U. S. A, 93:14164-14169 (1996).
J. Huwyler, J. Yang, and W. M. Pardridge. Receptor mediated delivery of
daunomycin using immunoliposomes: pharmacokinetics and tissue distribution in the
rat, J. Pharmacol Exp. Ther., 282:1541-1546 (1997).
H. linuma, K. Maruyama, K. Okinaga, K. Sasaki, T. Sekine, O. Ishida, N. Ogiwara,
K. Johkura, and Y. Yonemura. Intracellular targeting therapy of cisplatin-
encapsulated transferrin- polyethylene glycol liposome on peritoneal dissemination
of gastric cancer. Int. J. Cancer, 99:130-137 (2002).
B. E. Isakson, R. L. Lubman, G. J. Seedorf, and S. Boitano. Modulation of
pulmonary alveolar type II cell phenotype and communication by extracellular
matrix and KGF, Am. J. Physiol Cell Physiol, 281:C1291-C1299 (2001).
K. Iwai, S. K. Drake, N. B. Wehr, A. M. Weissman, T. LaVaute, N. Minato, R. D.
Klausner, R. L. Levine, and T. A. Rouault. Iron-dependent oxidation, ubiquitination,
and degradation of iron regulatory protein 2: implications for degradation of oxidized
proteins, Proc. Natl Acad. Set U. S. A, 95:4924-4928 (1998).
190
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
K. Iwai, R. D. Klausner, and T. A. Rouault. Requirements for iron-regulated
degradation of the RNA binding protein, iron regulatory protein 2, EMBO J.,
14:5350-5357 (1995).
S. Iyer and B. Lonnerdal. Lactoferrin, lactoferrin receptors and iron metabolism, Eur.
J. Clin. Nutr., 47:232-241 (1993).
P. H. Jarritt. Effect of iron on sedimentation-velocity and gel filtration behaviour of
transferrins from several vertebrates, Biochim. Biophys. Acta, 453:332-343 (1976).
W. A. Jefferies, M. R. Brandon, S. V. Hunt, A. F. Williams, K. C. Gatter, and D. Y.
Mason. Transferrin receptor on endothelium of brain capillaries. Nature, 312:162-
163 (1984).
P. D. Jeffrey, M. C. Bewley, R. T. MacGillivray, A. B. Mason, R. C. Woodworth,
and E. N. Baker. Ligand-induced conformational change in transferrins: crystal
structure of the open form of the N-terminal half-molecule of human transferrin.
Biochemistry, 37:13978-13986 (1998).
S. Q. Jing and I. S. Trowbridge. Identification of the intermolecular disulfide bonds
of the human transferrin receptor and its lipid-attachment site, EMBO J., 6:327-331
(1987).
C. Juhlin, S. Lundgren, H. Johansson, J. Lorentzen, L. Rask, E. Larsson, J. Rastad,
G. Akerstrom, and L. Klareskog. 500-Kilodalton calcium sensor regulating
cytoplasmic Ca2+ in cytotrophoblast cells of human placenta, /. Biol. Chem.,
265:8275-8279 (1990).
Katz J.H. Iron and protein kinetic studies by means of a doubly labeled human
crystalline transferrin., J. Clin. Invest., 40:2143-2152 (1961).
H. Kawabata, R. S. Germain, T. Ikezoe, X. Tong, E. M. Green, A. F. Gombart, and
H. P. Koeffler. Regulation of expression of murine transferrin receptor 2, Blood,
98:1949-1954 (2001).
191
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
H. Kawabata, R. S. Germain, P. T. Vuong, T. Nakamaki, J. W. Said, and H. P.
Koeffler. Transferrin receptor 2-alpha supports cell growth both in iron-chelated
cultured cells and in vivo, J. Biol. Chem., 275:16618-16625 (2000).
H. Kawabata, T. Nakamaki, P. Dconomi, R. D. Smith, R. S. Germain, and H. P.
Koeffler. Expression of transferrin receptor 2 in normal and neoplastic hematopoietic
cells. Blood, 98 :2714-2719 (2001).
H. Kawabata, R. Yang, T. Hirama, P. T. Vuong, S. Kawano, A. F. Gombart, and H.
P. Koeffler. Molecular cloning of transferrin receptor 2. A new member of the
transferrin receptor-like family, J. Biol. Chem. ,20826-20832 (1999).
D. Kerjaschki and M. G. Farquhar. The pathogenic antigen of Heymann nephritis is a
membrane glycoprotein of the renal proximal tubule brush border, Proc. Natl. Acad.
Sci. U. S. A, 79:5557-5581 (1982).
D. Kerjaschki, L. Noronha-Blob, B. Sacktor, and M. G. Farquhar. Microdomains of
distinctive glycoprotein composition in the kidney proximal tubule brush border, J.
Cell Biol., 98 :1505-1513 (1984).
R. Kircheis, S. Schuller, S. Brunner, M. Ogris, K. H. Heider, W. Zauner, and E.
Wagner. Polycation-based DNA complexes for tumor-targeted gene delivery in vivo,
J. Gene Med., 1:111-120 (1999).
R. Kircheis, L. Wightman, A. Schreiber, B. Robitza, Y. Rossler, M. Kursa, and E.
Wagner. Polyethylenimine/DNA complexes shielded by transferrin target gene
expression to tumors after systemic application. Gene Ther., 8:28-40 (2001).
R. D. Klausner, G. Ashwell, J. Van Renswoude, J. B. Harford, and K. R. Bridges.
Binding of apotransferrin to K562 cells: explanation of the transferrin cycle, Proc.
Natl. Acad. Sci. U. S. A, 80:2263-2266 (1983).
R. D. Klausner, G. Ashwell, J. van Renswoude, J. B. Harford, and K. R. Bridges.
Binding of apotransferrin to K562 cells: explanation of the transferrin cycle, Proc.
Natl. Acad. Sci. U. S. A, 80:2263-2266 (1983).
192
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
R. D. Klausner, J. G. Donaldson, and J. Lippincott-Schuartz. Brefeldin A: Insights
into the control of membrane traffic and orgenelle structure., J. Cell Biol., 116:1071-
1080 (1992).
R. D. Klausner, J. Van Renswoude, G. Ashwell, C. Kempf, A. N. Schechter, A.
Dean, and K. R. Bridges. Receptor-mediated endocytosis of transferrin in K562
cells, /. Biol. Chem., 258:4715-4724 (1983).
K. W. Ko, R. K. Avramoglu, R. S. McLeod, J. Vukmirica, and Z. Yao. The insulin-
stimulated cell surface presentation of low density lipoprotein receptor-related
protein in 3T3-L1 adipocytes is sensitive to phosphatidylinositide 3-kinase
inhibition. Biochemistry, 40:752-759 (2001).
K. Kono, Y. Torikoshi, M. Mitsutomi, T. Itoh, N. Emi, H. Yanagie, and T.
Takagishi. Novel gene delivery systems: complexes of fusigenic polymer-modified
liposomes and lipoplexes, Gene Ther., 8:5-12 (2001).
J. H. Kordower, V. Charles, R. Bayer, R. T. Bartus, S. Putney, L. R. Walus, and P.
M. Friden. Intravenous administration of a transferrin receptor antibody-nerve
growth factor conjugate prevents the degeneration of cholinergic striatal neurons in a
model of Huntington disease, Proc. Natl. Acad. Sci. U. S. A, 91:9077-9080 (1994).
S. B. Komfeld, J. E. Leonard, M. D. Mullen, and R. Taetle. Assessment of ligand
effects in intracellular trafficking of ricin A chain using anti-ricin hybridomas.
Cancer Res., 51:1689-1693 (1991).
R. Kozyraki, J. Fyfe, P. J. Verroust, C. Jacobsen, A. Dautry-Varsat, J. Gburek, T. E.
Willnow, E. I. Christensen, and S. K. Moestrup. Megalin-dependent cubilin-
mediated endocytosis is a major pathway for the apical uptake of transferrin in
polarized epithelia, Proc. Natl. Acad. Sci. U. S. A, 98:12491-12496 (2001).
R. Kozyraki, M. Kristiansen, A. Silahtaroglu, C. Hansen, C. Jacobsen, N.
Tommerup, P. J. Verroust, and S. K. Moestrup. The human intrinsic factor-vitamin
B12 receptor, cubilin: molecular characterization and chromosomal mapping of the
gene to lOp within the autosomal recessive megaloblastic anemia (MGAl) region.
Blood, 91:3593-3600 (1998).
193
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
F. Kratz, U. Beyer, T. Roth, N. Tarasova, P. Collery, F. Lechenault, A. Cazabat, P.
Schumacher, C. Unger, and U. Falken. Transferrin conjugates of doxorabicin:
synthesis, characterization, cellular uptake, and in vitro efficacy, J. Pharm. Sci.,
87:338-346 (1998).
F. Kratz, T. Roth, I. Fichiner, P. Schumacher, H. H. Fiebig, and C. Unger. In vitro
and in vivo efficacy of acid-sensitive transferrin and albumin doxorubicin conjugates
in a human xenograft panel and in the MDA-MB-435 mamma carcinoma model, J.
Drug Target, 8:305-318 (2000).
M. Kristiansen, R. Kozyraki, C. Jacobsen, E. Nexo, P. J. Verroust, and S. K.
Moestrup. Molecular dissection of the intrinsic factor-vitamin B12 receptor, cubilin,
discloses regions important for membrane association and ligand binding, J. Biol.
Chem., 274:20540-20544 (1999).
Kuwabara T., Kato Y., Kobayashi S., Suzuki H., and Sugiyama Y. Nonlinear
pharmacokinetics of a recombinant human granulocyte colony-stimulating factor
derivative (nartograstim): species differences among rats, monkeys and humans., J.
Pharmacol. Exp. Ther., 271:1535-1543 (1994).
J. C. Kwok and D. R. Richardson. The iron metabolism of neoplastic cells:
alterations that facilitate proliferation?. Grit Rev. Oncol. HematoL, 42:65-78 (2002).
J. E. Lamb, F. Ray, J. H. Ward, J. P. Kushner, and J. Kaplan. Intemalization and
subcellular localization of transferrin and transferrin receptors in HeLa cells, J. Biol.
Chem., 258:8751-8758 (1983).
J. W. Larrick and P. Cress well. Modulation of cell surface iron transferrin receptors
by cellular density and state of activation, J. Supramol. Struct., 11:579-586 (1979).
D. W. Laske, R. J. Youle, and E. H. Oldfield. Tumor regression with regional
distribution of the targeted toxin TF- CRM 107 in patients with malignant brain
tumors, Nat. Med., 3:1362-1368 (1997).
194
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
D. C. Lawe, A. Chawla, E. Merithew, J. Dumas, W. Carrington, K. Fogarty, L.
Lifshitz, R. Tuft, D. Lambright, and S. Corvera. Sequential roles for
phosphatidylinositol 3-phosphate and RabS in tethering and fusion of early
endosomes via their interaction with EEAl, J. Biol. Chem., 277:8611-8617 (2002).
C. M. Lawrence, S. Ray, M. Babyonyshev, R. Galluser, D. W. Borhani, and S. C.
Harrison. Crystal structure of the ectodomain of human transferrin receptor. Science,
286:779-782 (1999).
A. Le Bivic, A. Quaroni, B. Nichols, and E. Rodriguez-Boulan. Biogenetic pathways
of plasma membrane proteins in Caco-2, a human intestinal epithelial cell line., J.
Cell Biol., 111:1351-1361 (1990).
A. H. Le Gall, C. Yeaman, A. Muesch, and E. Rodriguez-Boulan. Epithelial cell
polarity: new perspectives., Seminars in Nephrology, 15:272-284 (1995).
J. A. Lebron, M. J. Bennett, D. E. Vaughn, A. J. Chirino, P. M. Snow, G. A. Mintier,
J. N. Feder, and P. J. Bjorkman. Crystal structure of the hemochromatosis protein
HFE and characterization of its interaction with transferrin receptor. Cell, 93:111-
123 (1998).
J. A. Lebron, A. P. West, Jr., and P. J. Bjorkman. The hemochromatosis protein HFE
competes with transferrin for binding to the transferrin receptor, J. Mol. Biol.,
294:239-245 (1999).
E. A. Leibold and H. N. Munro. Cytoplasmic protein binds in vitro to a highly
conserved sequence in the 5' untranslated region of ferritin heavy- and light-subunit
mRNAs, Proc. Natl. Acad. Sci. U. S. A, 85:2171-2175 (1988).
J. S. Levine, R. H. Allen, D. H. Alpers, and B. Seetharam. Immunocytochemical
localization of the intrinsic factor-cobalamin receptor in dog-ileum: distribution of
intracellular receptor during cell maturation, J. Cell Biol., 98:1111-1118 (1984).
J. E. Levy, L. K. Montross, D. E. Cohen, M. D. Fleming, and N. C. Andrews. The
C282Y mutation causing hereditary hemochromatosis does not produce a null allele.
Blood, 94:9-11 (1999).
195
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
O. Levy. Antibiotic proteins of polymorphonuclear leukocytes, Eur. J. Haematol,
56:263-277 (1996).
G. Li, C. D'Souza-Schorey, M. A. Barbieri, R. L. Roberts, A. Klippel, L. T.
Williams, and P. D. Stahl. Evidence for phosphatidylinositol 3-kinase as a regulator
of endocytosis via activation of RabS, Proc. Natl Acad. Sci U. S. A, 92:10207-
10211 (1995).
H. Li, P. J. Sadler, and H. Sun. Rationalization of the strength of metal binding to
human serum transferrin, Eur. J. Biochem., 242:387-393 (1996).
L. Li and J. Kaplan. Alteration in the organ distribution of iron by truncated
transferrin: implications for iron chelation therapy, J. Laboratory & Clinical
Medicine, 130:271-277 (1997).
Y. Li, T. Smith, S. Grabski, and D. L. DeWitt. The membrane association sequences
of the prostaglandin endoperoxide synthases-1 and -2 isozymes, J. Biol Chem.,
273:29830-29837 (1998).
A. J. Lindsay, A. G. Hendrick, G. Cantalupo, F. Senic-Matuglia, B. Goud, C. Bucci,
and M. W. McCaffrey. Rab coupling protein (RCP), a novel Rab4 and Rabl 1
effector protein, J. Biol Chem., 277:12190-12199 (2002).
A. J. Lindsay and M. W. McCaffrey. Rabll-FIP2 functions in transferrin recycling
and associates with endosomal membranes via its COOH-terminal domain, J. Biol
Chem., 277:27193-27199 (2002).
J. Lippincott-Schuartz, J. G. Donaldson, A. J. G. Schweizer, E. G. Berger, H. P.
Kauri, L. C. Yuan, and R. D. Klausner. Microtube-dependent retrograde transport of
proteins into the ER in the presence of brefeldin A suggests an ER recycling
pathway.. Cell, 60:821-836 (1990).
J. Lippincott-Schuartz, L. Yuan, C. Tipper, M. Amherdt, L. Orci, and R. D.
Klausner. Brefeldin A's effects on endosomes, lysosomes and TGN suggest a gemal
mechanism for regulating organelle structure and membrane traffic.. Cell, 67:601-
616(1991).
196
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
J. M. Lloyd, T. O'Dowd, M. Driver, and D. E. Tee. Demonstration of an epitope of
the transferrin receptor in human cervical epithelium-a potentially useful cell
marker, J. Clin. Pathol, 37:131-135 (1984).
B. Lonnerdal and S. Iyer. Lactoferrin: molecular structure and biological function,
Annu. Rev. Nutr., 15:93-110 (1995).
B. I. Lord, L. B. Woolford, and G. Molineux. Kinetics of neutrophil production in
normal and neutropenic animals during the response to filgrastim (r-metHu G-CSF)
or filgrastim SD/01 (PEG-r-metHu G-CSF), Clin. Cancer Res., 7:2085-2090 (2001).
S. H. Low, B. L. Tang, S. H. Wong, and W. J. Hong. Selective inhibition of protein
targeting to the apical domain of MDCK cells by brefeldin A., J. Cell Biol, 118:51-
62 (1992).
R. T. MacGillivray, S. A. Moore, J. Chen, B. F. Anderson, H. Baker, Y. Luo, M.
Bewley, C. A. Smith, M. E. Murphy, Y. Wang, A. B. Mason, R. C. Woodworth, G.
D. Brayer, and E. N. Baker. Two high-resolution crystal structures of the
recombinant N-lobe of human transferrin reveal a structural change implicated in
iron release. Biochemistry, 37:7919-7928 (1998).
D. G. Makey and U. S. Seal. The detection of four molecular forms of human
transferrin during the iron binding process, Biochim. Biophys. Acta, 453:250-256
(1976).
J. L. Martys, C. Wjasow, D. M. Gangi, M. C. Kielian, T. E. McGraw, and J. M.
Backer. Wortmarmin-sensitive trafficking pathways in Chinese hamster ovary cells.
Differential effects on endocytosis and lysosomal sorting, J. Biol. Chem., 271:10953-
10962 (1996).
A. B. Mason, B. M. Tam, R. C. Woodworth, R. W. Oliver, B. N. Green, L. N. Lin, J.
F. Brandts, K. J. Savage, J. A. Lineback, and R. T. MacGillivray. Receptor
recognition sites reside in both lobes of human serum transferrin, Biochem. J., 326 (
Pt l):77-85 (1997).
197
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
K. Matsushita, P. B. McCray, Jr., R. D, Sigmund, M. J. Welsh, and J. B. Stokes.
Localization of epithelial sodium channel subunit mRNAs in adult rat lung by in situ
hybridization. Am. J. Physiol, 271:L332-L339 (1996).
K. Matter, M. Brauchbar, K. Bucher, and H. P. Hauri. Sorting of endogenous plasma
membrane proteins occurs from two sites in cutured human intestinal epithelial cells
(Caco-2)., Cell, 60:429-437 (1990).
K. Matter, J. A. Whitney, E. M. Yamamoto, and I. Mellman. Common signals
control low density lipoprotein receptor sorting in endosomes and the Golgi complex
of MDCK cells. Cell, 74:1053-1064 (1993).
H. M. McBride, V. Rybin, C. Murphy, A. Giner, R. Teasdale, and M. Zerial.
Oligomeric complexes link RabS effectors with NSF and drive membrane fusion via
interactions between EEAl and syntaxin 13, Cell, 98:377-386 (1999).
D. J. McCool, J. F. Forstner, and G. G. Forstner. Regulated and unregulated
pathways for MUC2 mucin secretion in human colonic LS180 adenocarcinoma cells
are distinct, Biochem. J., 312 ( Pt 1): 125-133 (1995).
C. Mercier, M. F. Cesbron-Delauw, and L. D. Sibley. The amphipathic alpha helices
of the toxoplasma protein GRA2 mediate post-secretory membrane association, J.
Cell Sci., I l l ( Pt 15):2171-2180 (1998).
R. Meyers and L. C. Cantley. Cloning and characterization of a wortmannin-
sensitive human phosphatidylinositol 4-kinase, J. Biol. Chem., 272:4384-4390
(1997).
V. K. Mishra and M. N. Palgunachari. Interaction of model class A l, class A2, and
class Y amphipathic helical peptides with membranes, Biochemistry, 35:11210-
11220(1996).
A. Misumi, A. Miki, A. Takatsuki, G. Tamura, and Y. Ikehara. Novel blockade by
brefeldin A of intracellular transport of secretory proteins in rat hepacytes., J. Biol.
Chem., 261:11398-11403 (1986).
198
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
s. K. Moestrup, R. Kozyraki, M. Kristiansen, J. H. Kaysen, H. H. Rasmussen, D.
Brault, F. Pontillon, F. O. Goda, E. I. Christensen, T. G. Hammond, and P. J.
Verroust. The intrinsic factor-vitamin B12 receptor and target of teratogenic
antibodies is a megalin-binding peripheral membrane protein with homology to
developmental proteins, J. Biol. Chem., 273:5235-5242 (1998).
S. K. Moestrup and P. J. Verroust. Megalin- and cubilin-mediated endocytosis of
protein-bound vitamins, lipids, and hormones in polarized epithelia, Annu. Rev.
Nutr., 21:407-428 (2001).
K. Mohrmann, L. Gerez, V. Oorschot, J. Klumperman, and S. P. van der. Rab4
function in membrane recycling from early endosomes depends on a membrane to
cytoplasm cycle, J. Biol. Chem., 277:32029-32035 (2002).
K. Mohrmann, R. Leijendekker, L. Gerez, and S. P. van der. rab4 regulates transport
to the apical plasma membrane in Madin-Darby canine kidney cells, J. Biol. Chem.,
277:10474-10481 (2002).
T. Moos and E. H. Morgan. Restricted transport of anti-transferrin receptor antibody
(0X26) through the blood-brain barrier in the rat, J. Neurochem., 79:119-129
(2001).
A. G. Morell, G. Gregoriadis, I. H. Scheinberg, J. Hickman, and G. Ashwell. The
role of sialic acid in determining the survival of glycoproteins in the circulation, J.
Biol. Chem., 246:1461-1467 (1971).
E. H. Morgan, G. Marsaglia, E. R. Giblett, and C. A. Finch. A method of
investigating internal iron exchange utilizing two types of transferrin, J. Lab Clin.
Med., 69:370-381 (1967).
E. H. Morgan and T. Peters, Jr. Intracellular aspects of transferrin synthesis and
secretion in the rat, J. Biol. Chem., 246:3508-3511 (1971).
E. H. Morgan and T. Peters, Jr. The biosynthesis of rat serum albumin. V. Effect of
protein depletion and refeeding on albumin and transferrin synthesis, J. Biol. Chem.,
246:3500-3507 (1971).
199
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
A. G. Morton and A. S. TavilL The role of iron in the regulation of hepatic
transferrin synthesis, Br. J. Haematol, 36:383-394 (1977).
T. Mosmann. Rapid colorimetric assay for cellular growth and survival: application
to proliferation and cytotoxicity assays, J. Immunol Methods, 65:55-63 (1983).
K. E. Mostov and M. H. Cardone. Regulation of protein traffic in polarized epithelial
cells, Bioessays, 17:129-138 (1995).
F. T. Mu, J. M. Callaghan, O. Steele-Mortimer, H. Stenmark, R. G. Parton, P. L.
Campbell, J. McCluskey, J. P. Yeo, E. P. Tock, and B. H. Toh. EEAl, an early
endosome-associated protein. EEAl is a conserved alpha- helical peripheral
membrane protein flanked by cysteine "fingers" and contains a calmodulin-binding
IQ motif, J. Biol Chem., 270:13503-13511 (1995).
E. W. MuUner and L. C. Kuhn. A stem-loop in the 3' untranslated region mediates
iron-dependent regulation of transferrin receptor mRNA stability in the cytoplasm,
Cell, 53:815-825 (1988).
J. Narahari, R. Ma, M. Wang, and W. E. Walden. The aconitase function of iron
regulatory protein 1. Genetic studies in yeast implicate its role in iron-mediated
redox regulation, J. Biol Chem., 275:16227-16234 (2000).
B. Nikinmaa, C. A. Enns, S. E. Tonik, H. H. Sussman, and J. Schroder. Monoclonal
antibodies to a purified human transferrin receptor, Scand. J. Immunol, 20:441-447
(1984).
H. O'Brodovich, C. Canessa, J. Ueda, B. Rafii, B. C. Rossier, and J. Edelson.
Expression of the epithelial Na+ channel in the developing rat lung, Am. J. Physiol,
265:C491-C496 (1993).
K. Oda, S. Hirose, N. Takami, A. Misumi, A. Takatsuki, and Y. Ikehara. Brefedin A
arrests the intracellular transport of a precursor of completement C3 before its
converson site in rat hepatocytes., FEBS Lett., 214:135-138 (1987).
2 0 0
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
G. Odorizzi, A. Pearse, D. Domingo, I. S. Trowbridge, and C. R. Hopkins. Apical
and basolateral endosomes of MDCK cells are interconnected and contain a
polarized sorting mechanism, J. Cell Biol, 135:139-152 (1996).
O. Olakanmi, S. E. McGowan, M. B. Hayek, and B. E. Britigan. Iron sequestration
by macrophages decreases the potential for extracellular hydroxyl radical formation,
J. Clin. Invest, 91:889-899 (1993).
M. B. Omary and I. S. Trowbridge. Covalent binding of fatty acid to the transferrin
receptor in cultured human cells, J. Biol Chem., 256:4715-4718 (1981).
E. R. Pacht and W. B. Davis. Role of transferrin and ceruloplasmin in antioxidant
activity of lung epithelial lining fluid, J. Appl. Physiol, 64:2092-2099 (1988).
R. M. Palmour and H. E. Sutton. Vertebrae transferrins. Molecular weights, chemical
compositions, and iron-binding studies. Biochemistry, 10:4026-4032 (1971).
W. M. Pardridge, J. L. Buciak, and P. M. Friden. Selective transport of an anti
transferrin receptor antibody through the blood-brain barrier in vivo, J. Pharmacol.
Exp. Ther., 259:66-70 (1991).
W. M. Pardridge, J. L. Buciak, and P. M. Friden. Selective transport of an anti
transferrin receptor antibody through the blood-brain barrier in vivo, J. Pharmacol.
Exp. Ther., 259:66-70 (1991).
W. M. Pardridge, D. Wu, and T. Sakane. Combined use of carboxyl-directed protein
pegylation and vector- mediated blood-brain barrier drug delivery system optimizes
brain uptake of brain-derived neurotrophic factor following intravenous
administration, Pharm. Res., 15:576-582 (1998).
S. Parkkila, A. Waheed, R. S. Britton, B. R. Bacon, X. Y. Zhou, S. Tomatsu, R. E.
Fleming, and W. S. Sly. Association of the transferrin receptor in human placenta
with HFE, the protein defective in hereditary hemochromatosis, Proc. Natl Acad.
Sci U. S. A, 94:13198-13202 (1997).
2 0 1
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
s. Parkkila, A. Waheed, R. S. Britton, J. N. Feder, Z. Tsuchihashi, R. C . Schatzman,
B. R. Bacon, and W. S. Sly. Immunohistochemistry of HLA-H, the protein defective
in patients with hereditary hemochromatosis, reveals unique pattern of expression in
gastrointestinal tract, Proc. Natl. Acad. Sci. U. S. A, 94:2534-2539 (1997).
R. G. Parton. Endocytosis in polarized cells, Semin. Cell Biol., 2:387-395 (1991).
V. Patki, D. C. Lawe, S. Corvera, J. V. Virbasius, and A. Chawla. A functional
PtdIns(3)P-binding motif, Nature, 394:433-434 (1998).
R. Prekeris, J. Klumperman, and R. H. ScheUer. A Rabl 1/Rip 11 protein complex
regulates apical membrane trafficking via recycling endosomes. Mol. Cell, 6:1437-
1448 (2000).
A. C. Prost, F. Menegaux, P. Langlois, J. M. Vidal, M. Koulibaly, J. L. Jost, J. J.
Duron, J. P. Chigot, P. Vayre, A. Aurengo, J. C. Legrand, G. Rosselin, and C.
Gespach. Differential transferrin receptor density in human colorectal cancer: A
potential probe for diagnosis and therapy. Int. J. Oncol., 13:871-875 (1998).
K. Prydz, S. H. Hansen, K. Sandvig, and B. Van Deurs. Effects of brefeldin A on
endocytosis, transcytosis and transport to the Golgi complex in polarized MDCk
cells., J. Cell Biol, 119:259-272 (1992).
A. Quaroni and J. Hochman. Development of intestinal cell culture models for drug
transport and metabolism studies., Adv. Drug Delivery Rev., 22:3-52 (1996).
G. Quick, J. van Zyl, A. Hawtrey, and M. Ariatti. Effect of nicotinic acid conjugated
to DNA-transfecting complexes targeted at the transferrin receptor of HeLa cells,
DrugDeliv., 7:231-236 (2000).
H. N. Raaf, D. W. Jacobsen, S. Savon, and R. Green. Serum transferrin receptor
level is not altered in invasive adenocarcinoma of the breast, Am. J. Clin. Pathol,
99:232-237 (1993).
202
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
T. S. Ramalingam, A. P. West, Jr., J. A. Lebron, J. S. Nangiana, T. H. Hogan, C. A.
Enns, and P. J. Bjorkman. Binding to the transferrin receptor is required for
endocytosis of HFE and regulation of iron homeostasis, Nat. Cell Biol, 2:953-957
(2000).
K. S. Ramanujam, S. Seetharam, N. M. Dahms, and B. Seetharam. Effect of
processing inhibitors on cobalamin (vitamin B 12) transcytosis in polarized opossum
kidney cells. Arch. Biochem. Biophys., 315:8-15 (1994).
K. Rao, J. B. Harford, T. Rouault, A. McClelland, F. H. Ruddle, and R. D. Klausner.
Transcriptional regulation by iron of the gene for the transferrin receptor. Mol. Cell
Biol, 6:236-240 (1986).
R. Raychowdhury, J. L. Niles, R. T. McCluskey, and J. A. Smith. Autoimmune
target in Heymann nephritis is a glycoprotein with homology to the LDL receptor.
Science, 244:1163-1165 (1989).
L. D. Recht, T. W. Griffin, V. Raso, and A. R. Salimi. Potent cytotoxicity of an
antihuman transferrin receptor-ricin A-chain immunotoxin on human glioma cells in
vitro. Cancer Res., 50:6696-6700 (1990).
M. Ren, G. Xu, J. Zeng, C. Lemos-Chiarandini, M. Adesnik, and D. D. Sabatini.
Hydrolysis of GTP on rabl 1 is required for the direct delivery of transferrin from the
pericentiiolar recychng compartment to the cell surface but not from sorting
endosomes, Proc. Natl Acad. Sci U. S. A , 95:6187-6192 (1998).
D. R. Richardson and P. Ponka. The molecular mechanisms of the metabolism and
transport of iron in normal and neoplastic cells, Biochim. Biophys. Acta, 1331:1-40
(1997).
H. D. Riedel, M. U. Muckenthaler, S. G. Gehrke, I. Mohr, K. Brennan, T. Herrmann,
B. A. Fitscher, M. W. Hentze, and W. Stremmel. HFE downregulates iron uptake
from transferrin and induces iron- regulatory protein activity in stably transfected
cells. Blood, 94:3915-3921 (1999).
203
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
A. Roetto, F. Daraio, F. Alberti, P. Porporato, A. Cali, M. De Gobbi, and C.
Camaschella. Hemochromatosis due to mutations in transferrin receptor 2, Blood
Cells Mol. Dis., 29:465-470 (2002).
A. Roetto, A. Totaro, A. Pipemo, A. Piga, F. Longo, G. Garozzo, A. Cali, M. De
Gobbi, P. Gasparini, and C. Camaschella. New mutations inactivating transferrin
receptor 2 in hemochromatosis type 3, Blood, 97:2555-2560 (2001).
P. Rossi, F. Giansanti, A. Boffi, M. Ajello, P. Valenti, E. Chiancone, and G.
Antonini. Ca2-i- binding to bovine lactoferrin enhances protein stability and
influences the release of bacterial lipopolysaccharide, Biochem. Cell Biol, 80:41-48
(2002).
J. E. Rothman and G. Warren. Implications of the SNARE hypothesis for
intracellular membrane topology and dynamics, Curr. Biol, 4:220-233 (1994).
T. Rouault and R. Klausner. Regulation of iron metabolism in eukaryotes, Curr. Top.
Ce//i?eg«/., 35:1-19 (1997).
C. N. Roy and C. A. Enns. Iron homeostasis: new tales from the crypt. Blood,
96:4020-4027 (2000).
C. N. Roy, D. M. Penny, J. N. Feder, and C. A. Enns. The hereditary
hemochromatosis protein, HFE, specifically regulates transferrin-mediated iron
uptake in HeLa cells, J. Biol Chem., 274:9022-9028 (1999).
E. A. Rutledge and C. A. Enns. Cleavage of the transferrin receptor is influenced by
the composition of the O-linked carbohydrate at position 104, J. Cell Physiol,
168:284-293 (1996).
D. Sahali, N. Mulliez, F. Chatelet, R. Dupuis, P. Ronco, and P. Verroust.
Characterization of a 280-kD protein restricted to the coated pits of the renal brush
border and the epithelial cells of the yolk sac. Teratogenic effect of the specific
monoclonal antibodies, J. Exp. Med., 167:213-218 (1988).
204
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
D. Sahali, N. Mulliez, F. Chatelet, C. Laurent-Winter, D. Citadelle, C. Roux, P.
Ronco, and P. Verroust. Coexpression in humans by kidney and fetal envelopes of a
280 kDa- coated pit-restricted protein. Similarity with the murine target of
teratogenic antibodies, Am. J. Pathol., 140:33-44 (1992).
A. Saito, S. Pietromonaco, A. K. Loo, and M. G. Farquhar. Complete cloning and
sequencing of rat gp330/"megalin," a distinctive member of the low density
lipoprotein receptor gene family, Proc. Natl. Acad. Sci. U. S. A, 91:9725-9729
(1994).
M. R. Schlabach and G. W. Bates. The synergistic binding of anions and Fe3+ by
transferrin. Implications for the interlocking sites hypothesis, J. Biol. Chem.,
250:2182-2188 (1975).
B. Schlierf, G. H. Fey, J. Hauber, G. M. Hocke, and O. Rosorius. Rabl lb is essential
for recycling of transferrin to the plasma membrane. Exp. Cell Res., 259:257-265
(2000).
H. M. Schulman, J. Martinez-Medellin, and R. Sidloi. The reticulocyte-mediated
release of iron and bicarbonate from transferrin: effect of metabolic inhibitors,
Biochim. Biophys. Acta, 343:529-534 (1974).
B. Seetharam, E. I. Christensen, S. K. Moestrup, T. G. Hammond, and P. J. Verroust.
Identification of rat yolk sac target protein of teratogenic antibodies, gp280, as
intrinsic factor-cobalamin receptor, J. Clin. Invest, 99:2317-2322 (1997).
B. Seetharam, J. S. Levine, M. Ramasamy, and D. H. Alpers. Purification,
properties, and immunochemical localization of a receptor for intrinsic factor-
cobalamin complex in the rat kidney, J. Biol. Chem., 263:4443-4449 (1988).
M. Seki, J. Iwakawa, H. Cheng, and P. W. Cheng. p53 and PTEN/MMAC1/TEP1
gene therapy of human prostate PC-3 carcinoma xenograft, using transferrin-
facilitated lipofection gene delivery strategy, Hum. Gene Ther., 13:761-773 (2002).
205
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
E. Sekyere and D. R. Richardson. The membrane-bound transferrin homologue
melanotransferrin: roles other than iron transport?, FEBS Lett., 483:11-16 (2000).
D. Shah and W. C. Shen. The establishment of polarity and enhancd transcytosis of
transferrin receptors in enterocyte-like Caco-2 cells., J. Drug Targeting, 2:93-99
(1994).
D. Shah and W. C. Shen. Transcellular delivery of an insulin-transferrin conjugate in
enterocyte-like Caco-2 cells, J. Pharm. Sci., 85:1306-1311 (1996).
J. M. Shannon, S. D. Jennings, and L. D. Nielsen. Modulation of alveolar type II cell
differentiated function in vitro, Am. J. Physiol, 262:L427-L436 (1992).
D. Sheff, L. Pelletier, C. B. O'Connell, G. Warren, and I Mellman. Transferrin
receptor recycling in the absence of perinuclear recycling endosomes, J. Cell Biol,
156:797-804 (2002).
D. R. Sheff, E. A. Daro, M. Hull, and I. Mellman. The receptor recycling pathway
contains two distinct populations of early endosomes with different sorting functions,
J. Cell Biol, 145:123-139 (1999).
D. R. Sheff, R. Kroschewski, and I. Mellman. Actin dependence of polarized
receptor recycling in Madin-Darby canine kidney cell endosomes. M ol Biol Cell,
13:262-275 (2002).
W. C. Shen, D. Shah, M. Taub, and J. Wan. Enhancement of transferrin receptor-
mediated transcytosis for transepithelial protein delivery. In V. H. L. Lee, M.
Hashida, and Y. Mizushima (eds). Trends and future perspectives in peptide and
protein drug delivery., harwood academic publishers, 1995, pp. 135-151.
P. R. Shepherd, M. A. Soos, and K. Siddle. Inhibitors of phosphoinositide 3-kinase
block exocytosis but not endocytosis of transferrin receptors in 3T3-L1 adipocytes,
Biochem. Biophys. Res. Commun., 211:535-539 (1995).
N. Shi, Y. Zhang, C. Zhu, R. J. Boado, and W. M. Pardridge. Brain-specific
expression of an exogenous gene after i.v. administration, Proc. Natl Acad. Sci U.
S. A, 98:12754-12759 (2001).
206
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
J. E. Shindelman, A. E. Ortmeyer, and H. H. Sussman. Demonstration of the
transferrin receptor in human breast cancer tissue. Potential marker for identifying
dividing cells. Int. J. Cancer, 27:329-334 (1981).
N. Shirafuji, S. Asano, S. Matsuda, K. Watari, F. Takaku, and S. Nagata. A new
bioassay for human granulocyte colony-stimulating factor (hG-CSF) using murine
myeloblastic NFS-60 cells as targets and estimation of its levels in sera from normal
healthy persons and patients with infectious and hematological disorders, Exp.
HematoL, 17:116-119 (1989).
F[. Shpetner, M. Joly, D. Hartley, and S. Corvera. Potential sites of PI-3 kinase
function in the endocytic pathway revealed by the PI-3 kinase inhibitor, wortmannin,
J. Cell Biol, 132:595-605 (1996).
U. Siddhanta, J. McDroy, A. Shah, Y. Zhang, and J. M. Backer. Distinct roles for the
pi lOalpha and hVPS34 phosphatidylinositol 3'- kinases in vesicular trafficking,
regulation of the actin cytoskeleton, and mitogenesis, J. Cell Biol., 143:1647-1659
(1998).
S. Simoes, P. Pires, N. Duzgunes, and M. C. Pedrosa de Lima. Cationic liposomes as
gene transfer vectors: barriers to successful application in gene therapy, Curr. Opin.
Mol. Ther., 1:147-157 (1999).
S. Simoes, V. Slepushkin, R. Caspar, M. C. de Lima, and N. Duzgunes. Gene
delivery by negatively charged ternary complexes of DNA, cationic liposomes and
transferrin or fusigenic peptides, Gene Ther., 5:955-964 (1998).
K. Simons and A. Wandinger-Ness. Polarized sorting in epithelia.. Cell, 62:207-210
(1990).
A. Simonsen, J. M. Gaullier, A. D'Arrigo, and H. Stenmark. The Rab5 effector
EEAl interacts directly with syntaxin-6, J. Biol. Chem., 274:28857-28860 (1999).
A. Simonsen, R. Lippe, S. Christoforidis, J. M. Gaullier, A. Brech, J. Callaghan, B.
H. Toh, C. Murphy, M. Zerial, and H. Stenmark. EEAl links PI(3)K function to
Rab5 regulation of endosome fusion. Nature , 394:494-498 (1998).
207
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
M. Singh. Transferrin As A targeting ligand for liposomes and anticancer drugs,
Curr. Pharm. Des, 5:443-451 (1999).
M. Singh, H. Atwal, and R. Micetich. Transferrin directed delivery of adriamycin to
human cells. Anticancer Res., 18:1423-1427 (1998).
K. R. Smith and A. E. Aust. Mobilization of iron from urban particulates leads to
generation of reactive oxygen species in vitro and induction of ferritin synthesis in
human lung epithelial cells, Chem. Res. Toxicol, 10:828-834 (1997).
B. W. Song, H. V. Vinters, D. Wu, and W. M. Pardridge. Enhanced neuroprotective
effects of basic fibroblast growth factor in regional brain ischemia after conjugation
to a blood-brain barrier delivery vector, J. Pharmacol Exp. Ther., 301:605-610
(2002).
B. Sonnichsen, S. De Renzis, E. Nielsen, J. Rietdorf, and M. Zerial. Distinct
membrane domains on endosomes in the recycling pathway visualized by multicolor
imaging of Rab4, Rab5, and R abll, J. Cell B iol, 149:901-914 (2000).
S. Sonoda and M. Schlamowitz. Studies of 1251 trace labeling of immunoglobulin G
by chloramine-T, Immunochemistry., 7:885-898 (1970).
L. M. Souza, T. C. Boone, J. Gabiilove, P. H. Lai, K. M. Zsebo, D. C. Murdock, V.
R. Chazin, J. Bruszewski, H. Lu, K. K. Chen, and . Recombinant human granulocyte
colony-stimulating factor: effects on normal and leukemic myeloid cells. Science,
232:61-65 (1986).
G. Spik, B. Bayard, B. Foumet, G. Strecker, S. Bouquelet, and J. Montreuil. Studies
on glycoconjugates. LXIV. Complete structure of two carbohydrate units of human
serotransferrin, FEBS Lett., 50:296-299 (1975).
D. J. Spiro, W. Boll, T. Kirchhausen, and M. Wessling-Resnick. Wortmannin alters
the transferrin receptor endocytic pathway in vivo and in vitro. Mol. Biol Cell,
7:355-367 (1996).
208
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
B. S. Stein and H. H. Sussman. Demonstration of two distinct transferrin receptor
recycling pathways and transferrin-independent receptor internalization in K562
cells, J. Biol. Chem., 261:10319-10331 (1986).
M. Steiner. Fluorescence microphotometric studies of the transferrin receptor in
human erythroid precursor cells, J. Lab Clin. Med., 96:1086-1093 (1980).
H. Stenmark, R. Aasland, B. H. Toh, and A. D'Arrigo. Endosomal localization of the
autoantigen EEAl is mediated by a zinc- binding FYVE finger, J. Biol. Chem.,
271:24048-24054 (1996).
S. W. Stites, M. E. Nelson, and L. J. Wesselius. Transferrin concentrations in serum
and lower respiratory tract fluid of mechanically ventilated patients with COPD or
ARDS, Chest, 107:1681-1685 (1995).
K. Sugahara, J. S. Rubin, R. J. Mason, E. L. Aronsen, and J. M. Shannon.
Keratinocyte growth factor increases mRNAs for SP-A and SP-B in adult rat
alveolar type II cells in culture, Am. J. Physiol, 269:L344-L350 (1995).
H. Sun, M. C. Cox, H. Li, A. B. Mason, R. C. Woodworth, and P. J. Sadler.
[1H,13C] NMR determination of the order of lobe loading of human transferrin with
iron: comparison with other metal ions, FEBS Lett., 422:315-320 (1998).
H. Sun, H. Li, and P. J. Sadler. Transferrin as a metal ion mediator, Chem. Rev.,
99:2817-2842(1999).
R. Sutherland, D. Delia, C. Schneider, R. Newman, J. Kemshead, and M. Greaves.
Ubiquitous cell-surface glycoprotein on tumor cells is proliferation- associated
receptor for transferrin, Proc. Natl. Acad. Sci. U. S. A, 78:4515-4519 (1981).
Y. Takai, T. Sasaki, and T. Matozaki. Small GTP-binding proteins. Physiol Rev.,
81:153-208 (2001).
Tanaka H. and Kaneko T. Pharmacokinetics of recombinant human granulocyte
colony-stimulating factor in mice.. Blood, 79:536-539 (1992).
209
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
A. B. Thompson, T. Bohling, A. Heires, J. Linder, and S. I. Rennard. Lower
respiratory tract iron burden is increased in association with cigarette smoking, J.
Lab Clin. Med., 117:493-499 (1991).
P. E. Thorpe, P. Wallace, P. Knowles, M. Relf, A. Brown, G. Waston, D. Blakey,
and D. Newell. Improved antitumor effects of immunotoxins prepared with
deglycosylated ricin A-chain and hindered disulfide linkages, Cancer Res., 48:6396-
6403 (1988).
L. M. Timchak, F. Kruse, M. H. Marnell, and R. K. Draper. A thermosensitive lesion
in a Chinese hamster cell mutant causing differential effects on the acidification of
endosomes and lysosomes, J. Biol. Chem., 261:14154-14159 (1986).
A. G. Tonevitsky, I. A. Demina, 1.1. Agapov, G. V. Fattakhova, E. N. Popova, M.
M. Moysenovich, I. S. Komolov, A. S. Khalanskii, and M. P. Kirpichnikov.
Cytotoxic activities of conjugates of human transferrin and A subunits of plant toxins
both in vitro and in vivo, Dokl. Biochem., 374:210-213 (2000).
X. Tong, H. Kawabata, and H. P. Koeffler. Iron deficiency can upregulate expression
of transferrin receptor at both the mRNA and protein level, Br. J. Haematol,
116:458-464 (2002).
M. Trischler, W. Stoorvogel, and O. Ullrich. Biochemical analysis of distinct Rab5-
and Rabl 1-positive endosomes along the transferrin pathway, J. Cell Sci., 112 ( Ft
24):4773-4783 (1999).
d. Tros, I and N. Duzgunes. Efficient gene transfer by transferrin lipoplexes in the
presence of serum, Biochim. Biophys. Acta, 1463:333-342 (2000).
I. S. Trowbridge and F. Lopez. Monoclonal antibody to transferrin receptor blocks
transferrin binding and inhibits human tumor cell growth in vitro, Proc. Natl. Acad.
Sci. U. S. A, 79:1175-1179 (1982).
I. S. Trowbridge, R. A. Newman, D. L. Domingo, and C. Sauvage. Transferrin
receptors: structure and function, Biochem. Pharmacol, 33:925-932 (1984).
210
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
O. Ullrich, S. Reinsch, S. Urbe, M. Zerial, and R. G. Parton. Rabl 1 regulates
recycling through the pericentriolar recycling endosome, J. Cell Biol., 135:913-924
(1996).
J. B. Ulmer and G. E. Palade. Targeting and processing of glycophorins in murine
erythroleukemia cells: use of brefeldin A as a perturbant of intracellular traffic, Proc.
Natl. Acad. Sci. U. S. A, 86:6992-6996 (1989).
J. B. Ulmer and G. E. Palade. Effects of brefeldin A on the processing of viral
envelope glycoproteins in murine erythroleukemia cells, J. Biol. Chem., 266:9173-
9179(1991).
S. Urbe, L. A. Huber, M. Zerial, S. A. Tooze, and R. G. Parton. R abll, a small
GTPase associated with both constitutive and regulated secretory pathways in PC12
cells, FEBS Lett., 334:175-182 (1993).
E. M. van Dam and W. Stoorvogel. Dynamin-dependent transferrin receptor
recycling by endosome-derived clathrin-coated vesicles, Mol. Biol. Cell, 13:169-182
(2002).
E. M. van Dam, T. Ten Broeke, K. Jansen, P. Spijkers, and W. Stoorvogel.
Endocytosed transferrin receptors recycle via distinct dynamin and
phosphatidylinositol 3-kinase-dependent pathways, J. Biol. Chem. , 277:48876-
48883 (2002).
S. P. van der, M. Hull, L. A. Huber, P. Male, B. Goud, and I. Mellman. Reversible
phosphorylation— dephosphorylation determines the localization of rab4 during the
cell cycle, EMBO J., 11:4379-4389 (1992).
C. J. Vlahos, W. F. Matter, K. Y. Hui, and R. F. Brown. A specific inhibitor of
phosphatidylinositol 3-kinase, 2-(4- morpholinyl)-8-phenyl-4H-l-benzopyran-4-one
(LY294002), J. Biol. C/iem. ,5241-5248 (1994).
H. G. Wada, P. E. Hass, and H. H. Sussman. Transferrin receptor in human placental
brush border membranes. Studies on the binding of transferrin to placental
membrane vesicles and the identification of a placental brush border glycoprotein
with high affinity for transferrin, J. Biol. Chem., 254:12629-12635 (1979).
211
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
E. Wagner, M. Gotten, R. Foisner, and M. L. Bimstiel. Transferrin-polycation-DNA
complexes: the effect of polycations on the structure of the complex and DNA
delivery to cells, Proc. Natl. Acad. Sci. U. S. A, 88:4255-4259 (1991).
B. Wagner, K. Zatloukal, M. Gotten, H. Kirlappos, K. Mechtler, D. T. Guriel, and M.
L. Bimstiel. Goupling of adenoviras to transferrin-polylysine/DNA complexes
greatly enhances receptor-mediated gene delivery and expression of transfected
genes, Proc. Natl. Acad. Sci. U. S. A, 89:6099-6103 (1992).
E. Wagner, M. Zenke, M. Gotten, H. Beug, and M. L. Bimstiel. Transferrin-
polycation conjugates as carriers for DNA uptake into cells, Proc. Natl. Acad. Sci. U.
5. A, 87:3410-3414 (1990).
M. Wagner, A. K. Rajasekaran, D. K. Hanzel, S. Mayor, and E. Rodriguez-Boulan.
Brefeldin A causes stmctural and functional alterations of trans-Golgi network of
MDGK cells., J. Cell Set., 107:933-943 (1994).
A. Waheed, S. Parkkila, X. Y. Zhou, S. Tomatsu, Z. Tsuchihashi, J. N. Feder, R. G.
Schatzman, R. S. Britton, B. R. Bacon, and W. S. Sly. Hereditary hemochromatosis:
effects of G282Y and H63D mutations on association with beta2-microglobulin,
intracellular processing, and cell surface expression of the HFE protein in GOS-7
cells, Proc. Natl. Acad. Sci. U. S. A, 94:12384-12389 (1997).
D. M. Wallace, A. J. Lindsay, A. G. Hendrick, and M. W. McGaffrey. The novel
Rabll-FIP/Rip/RGP family of proteins displays extensive homo- and hetero
interacting abilities, Biochem. Biophys. Res. Commun., 292:909-915 (2002).
J. Wan, M. Taub, D. Shah, and W. G. Shen. Brefeldin A enhances receptor-mediated
transcytosis of Tf in filter grown Madin-darby canine kidney cells., J. Biol. Chem.,
267:13446-13450 (1992).
J. Wan, M. E. Taub, D. Shah, and W. G. Shen. Brefeldin A enhances receptor-
mediated transcytosis of transferrin in filter-grown Madin-Darby canine kidney cells,
J. Biol. Chem., 267:13446-13450 (1992).
212
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
E. Wang, P. S. Brown, B. Aroeti, S. J. Chapin, K. E. Mostov, and K. W. Dunn.
Apical and basolateral endocytic pathways of MDCK ceils meet in acidic common
endosomes distinct from a nearly-neutral apical recycling endosome. Traffic., 1:480-
493 (2000).
J. Wang and K. Pantopoulos. Conditional derepression of ferritin synthesis in cells
expressing a constitutive IRPl mutant, Mol. Cell Biol., 22:4638-4651 (2002).
J. Wang, D. Shen, and W. C. Shen. Oral delivery of insulin-transferrin conjugate in
streptozotocin-treated CF/1 mice, Pharm. Res. (Supplement), 14:469 (1997).
A. P. West, Jr., M. J. Bennett, V. M. Sellers, N. C. Andrews, C. A. Enns, and P. J.
Bjorkman. Comparison of the interactions of transferrin receptor and transferrin
receptor 2 with transferrin and the hereditary hemochromatosis protein HFE, J. Biol.
Chem., 275:38135-38138 (2000).
Widera A., Kim K.J., Crandall E.D., and Shen W.-C. Transcytosis of GCSF-
Transferrin Across Rat Alveolar Epithelial Cell Monolayers, Pharm. Res., 20: 1231-
1238 (2003).
Widera, A. W., Beloussow K., Kim K.J., Crandall E.D., and Shen W.C. Phenotype-
dependent synthesis of transferrin receptor in rat alveolar epithelial cell monolayers.
Cell Tissue Res., 312 (3): 313-318 (2003).
C. Wiedemann and S. Cockcroft. Vesicular transport. Sticky fingers grab a lipid.
Nature, 394:426-427 (1998).
A. M. Williams and C. A. Enns. A mutated transferrin receptor lacking asparagine-
linked glycosylation sites shows reduced functionality and an association with
binding immunoglobulin protein, J. Biol. Chem., 266:17648-17654 (1991).
T. E. Willnow, A. Nykjaer, and J. Herz. Lipoprotein receptors: new roles for ancient
proteins, Nat. Cell Biol., 1:E157-E162 (1999).
D. B. Wilson and M. P. Wilson. Identification and subcellular localization of human
rab5b, a new member of the ras-related superfamily of GTPases, J. Clin. Invest,
89:996-1005 (1992).
213
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
M. A. Winkler, J. O. Price, P. D. Foglesong, and W. H. West. Biodistribution and
plasma survival in mice of anti-melanoma monoclonal antibody cross-linked to
0KT3., Cancer Immunology, Immunotherapy., 31:278-284 (1990).
S. Wood, J. Park, and W. Brown. Brefedin A causes a microtuble-mediated fusion of
the TGN and early endosomes., Cell, 67:591-600 (1998).
D, Wu and W. M. Pardridge. Neuroprotection with noninvasive neurotrophin
delivery to the brain, Proc. Natl. Acad. Sci. U. S. A, 96:254-259 (1999).
D. Wu, B. W. Song, H. V. Vinters, and W. M. Pardridge. Pharmacokinetics and
brain uptake of biotinylated basic fibroblast growth factor conjugated to a blood-
brain barrier drug delivery system, J. Drug Target, 10:239-245 (2002).
C. Q. Xia and W. C. Shen. Tyrphostin-8 enhances transferrin receptor-mediated
transcytosis in Caco-2- cells and inreases hypoglycemic effect of orally administered
insulin-transferrin conjugate in diabetic rats, Pharm. Res., 18:191-195 (2001).
C. Q. Xia, J. Wang, and W. C. Shen. Hypoglycemic effect of insulin-transferrin
conjugate in streptozotocin- induced diabetic rats, J. Pharmacol. Exp. Ther.,
295:594-600 (2000).
D. Xu, R. Kozyraki, T. C. Newman, and J. C. Fyfe. Genetic evidence of an accessory
activity required specifically for cubilin brush-border expression and intrinsic factor-
cobalamin absorption. Blood, 94:3604-3606 (1999).
L. Xu, P. Frederik, K. F. Pirollo, W. H. Tang, A. Rait, L. M. Xiang, W. Huang, I.
Cruz, Y. Yin, and E. H. Chang. Self-assembly of a virus-rnirnicking nanostructure
system for efficient tumor-targeted gene delivery. Hum. Gene Ther., 13:469-481
(2002).
L. Xu, K. F. Pirollo, and E. H. Chang. Transferrin-liposome-mediated p53
sensitization of squamous cell carcinoma of the head and neck to radiation in vitro,
Hum. Gene Ther., 8:467-475 (1997).
214
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
L. Xu, K. F. Pirollo, and E. H. Chang. Tumor-targeted p53-gene therapy enhances
the efficacy of conventional chemo/radiotherapy, J. Control Release, 74:115-128
(2001).
L. Xu, K. F. Pirollo, W. H. Tang, A. Rait, and E. H. Chang. Transferrin-liposome-
mediated systemic p53 gene therapy in combination with radiation results in
regression of human head and neck cancer xenografts, Hum. Gene Ther., 10:2941-
2952 (1999).
K. Yanagihara, H. Cheng, and P. W. Cheng. Effects of epidermal growth factor,
transferrin, and insulin on lipofection efficiency in human lung carcinoma cells.
Cancer Gene Ther., 7:59-65 (2000).
Y. Yeh, S. Iwal, and R. E. Feeney. Conformations of denatured and renatured
ovotransferrin. Biochemistry, 18:882-889 (1979).
S. P. Young, A. Bomford, and R. Williams. The effect of the iron saturation of
transferrin on its binding and uptake by rabbit reticulocytes, Biochem. J., 219:505-
510(1984).
O. Zak, D. Trinder, and P. Aisen. Primary receptor-recognition site of human
transferrin is in the C- terminal lobe, .J . Biol. Chem., 269:7110-7114 (1994).
M. Zenke, P. Steinlein, E. Wagner, M. Cotten, H. Beug, and M. L. Bimstiel.
Receptor-mediated endocytosis of transferrin-polycation conjugates: an efficient way
to introduce DNA into hematopoietic cells, (1990).
Y. Zhang and W. M. Pardridge. Conjugation of brain-derived neurotrophic factor to
a blood-brain barrier drug targeting system enables neuroprotection in regional brain
ischemia following intravenous injection of the neurotrophin. Brain Res., 889:49-56
(2001).
Y. Zhang and W. M. Pardridge. Mediated efflux of IgG molecules from brain to
blood across the blood- brain barrier, J. Neuroimmunol., 114:168-172 (2001).
215
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Y. Zhang and W. M. Pardridge. Neuroprotection in transient focal brain ischemia
after delayed intravenous administration of brain-derived neurotrophic factor
conjugated to a blood-brain barrier drug targeting system, Stroke, 32:1378-1384
(2001).
Y. Zhang and W. M. Pai'dridge. Rapid transferrin efflux from brain to blood across
the blood-brain barrier, J. Neurochem., 76:1597-1600 (2001).
X. Y. Zhou, S. Tomatsu, R. E. Fleming, S. Parkkila, A. Waheed, J. Jiang, Y. Fei, E.
M. Brunt, D. A. Ruddy, C. E. Prass, R. C. Schatzman, R. O'Neill, R. S. Britton, B. R.
Bacon, and W. S. Sly. HFE gene knockout produces mouse model of hereditary
hemochromatosis, Proc. Natl. Acad. Sci. U. S. A, 95:2492-2497 (1998).
216
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
APPENDIX
N U M E R I C A L L Y O R D E R E D R E F E R E N C E S
1. P. Aisen and I. Listowsky. Iron transport and storage proteins, Annu. Rev.
Biochem., 49:357-393 (1980).
2. P. Aisen and E. B. Brown. The iron-binding function of transferrin in iron
metabolism, Semin. HematoL, 14:31-53 (1977).
3. P. Aisen and E. B. Brown. Structure and function of transferrin, Prog.
HematoL, 9:25-56 (1975).
4. E. N. Baker and P. F. Bindley. New perspectives on the structure and
function of transferrins, J. Inorg. Biochem., 47:147-160 (1992).
5. D. R. Richardson and P. Ponka. The molecular mechanisms of the
metabolism and transport of iron in normal and neoplastic cells, Biochim.
Biophys. Acta, 1331:1-40 (1997).
6. C. A. Enns and H. H. Sussman. Physical characterization of the transferrin
receptor in human placentae, J. Biol. Chem., 256:9820-9823 (1981).
7. C. A. Enns and H. H. Sussman. Similarities between the transferrin receptor
proteins on human reticulocytes and human placentae, J. Biol. Chem.,
256:12620-12623 (1981).
8. H. Huebers, E. Huebers, W. Rummel, and R. R. Crichton. Isolation and
characterization of iron-hinding proteins from rat intestinal mucosa, Eur. J.
Biochem., 66:447-455 (1976).
9. B. Bloch, T. Popovici, S. Chouham, M. J. Levin, D. Tuil, and A. Kahn.
Transferrin gene expression in choroid plexus of the adult rat brain. Brain
Res. Bull., 18:573-576 (1987).
217
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
10. B. Bloch, T. Popovici, M. J. Levin, D. Tuil, and A. Kahn. Transferrin gene
expression visualized in oligodendrocytes of the rat brain by using in situ
hybridization and immunohistochemistry, Proc. Natl. Acad. Sci. U. S. A,
82:6706-6710(1985).
11. Baggiolilni, A., Deduve, C., Masson, P. L., and Heremans, J. F. Association
of lactoferrin with specific granules in rabbit heterophil leucocytes.
J.Exp.Med. 131, 559-570 (1970).
12. D. R. Ambruso and R. B. Johnston, Jr. Lactoferrin enhances hydroxyl radical
production by human neutrophils, neutrophil particulate fractions, and an
enzymatic generating system, J. Clin. Invest, 67:352-360 (1981).
13. B. Lonnerdal and S. Iyer. Lactoferrin: molecular structure and biological
function, Annu. Rev. Nutr., 15:93-110 (1995).
14. O. Levy. Antibiotic proteins of polymorphonuclear leukocytes, Eur. J.
Haematol, 56:263-277 (1996).
15. S. Iyer and B. Lonnerdal. Lactoferrin, lactoferrin receptors and iron
metabolism, Eur. J. Clin. Nutr., 47:232-241 (1993).
16. J. H. Brock and I. Esparza. Failure of reticulocytes to take up iron from
lactoferrin saturated by various methods, Br. J. Haematol, 42:481-483
(1979).
17. P. Rossi, F. Giansanti, A. Boffi, M. Ajello, P. Valenti, E. Chiancone, and G.
Antonini. Ca2+ binding to bovine lactoferrin enhances protein stability and
influences the release of bacterial lipopolysaccharide, Biochem. Cell Biol,
80:41-48 (2002).
18. F. Giansanti, P. Rossi, M. T. Massucci, D. Botti, G. Antonini, P. Valenti, and
L. Seganti. Antiviral activity of ovotransferrin discloses an evolutionary
strategy for the defensive activities of lactoferrin, Biochem. Cell Biol,
80:125-130 (2002).
218
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
19. J. Gautron, M. T. Hincke, M . Panheleux, J. M. Garcia-Ruiz, T. Boldicke, and
Y. Nys. Ovotransferrin is a matrix protein of the hen eggshell membranes and
basal calcified layer, Connect. Tissue Res., 42:255-267 (2001).
20. M. R. Food, E. O. Sekyere, and D. R. Richardson. The soluble form of the
membrane-bound transferrin homologue, melanotransferrin, inefficiently
donates iron to cells via nonspecific internalization and degradation of the
protein, Eur. J. Biochem., 269:4435-4445 (2002).
21. E. Sekyere and D. R. Richardson. The membrane-bound transferrin
homologue melanotransferrin: roles other than iron transport?, FEBS Lett.,
483:11-16(2000).
22. J. P. Brown, R. M. Hewick, I. Hellstrom, K. E. Hellstrom, R. F. Doolittle,
and W. J. Dreyer. Human melanoma-associated antigen p97 is structurally
and functionally related to transferrin, Nature, 296:171-173 (1982).
23. J. C. Kwok and D. R. Richardson. The iron metabolism of neoplastic cells:
alterations that facilitate proliferation?, Grit Rev. Oncol. HematoL, 42:65-78
(2002).
24. I. Graham and J. Williams. A comparsion of glycopeptides from the
transferrins of several species, Biochem. J. , 145:263-279 (1975).
25. G. Spik, B. Bayard, B. Foumet, G. Strecker, S. Bouquelet, and J. Montreuil.
Studies on glycoconjugates. LXIV. Complete structure of two carbohydrate
units of human serotransferrin, FEBS Lett., 50:296-299 (1975).
26. C. J. Dekker, M. J. Kroos, H. C. Van der, and H. G. Van Eijk. Uptake of sialo
and asialo transferrins by isolated rat hepatocytes. Comparison of a
heterologous and a homologous system, Int. J. Biochem., 17:701-706 (1985).
27. E. H. Morgan, G. Marsaglia, E. R. Giblett, and C. A. Finch. A method of
investigating internal iron exchange utilizing two types of transferrin, J. Lab
Clin. Med., 69:370-381 (1967).
219
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
28. A. G. Morell, G. Gregoriadis, I. H. Scheinberg, J. Hickman, and G. Ashwell.
The role of sialic acid in determining the survival of glycoproteins in the
circulation, J. Biol. Chem., 246:1461-1467 (1971).
29. M. R. Schlabach and G. W. Bates. The synergistic binding of anions and
Fe3+ by transferrin. Implications for the interlocking sites hypothesis, J. Biol.
Chem., 250:2182-2188 (1975).
30. G. W. Bates and M. R. Schlabach. The nonspecific binding of Fe3+ to
transferrin in the absence of synergistic anions, J. Biol. Chem., 250:2177-
2181 (1975).
31. P. Aisen, A. Leibman, and R. A. Pinkowitz. The anion-binding functions of
transferrin, Adv. Exp. Med. B iol, 48:125-140 (1974).
32. P. D. Jeffrey, M. C. Bewley, R. T. MacGillivray, A. B. Mason, R. C.
Woodworth, and E. N. Baker. Ligand-induced conformational change in
transferrins: crystal structure of the open form of the N-terminal half
molecule of human transferrin. Biochemistry, 37:13978-13986 (1998).
33. R. T. MacGillivray, S. A. Moore, J. Chen, B. F. Anderson, H. Baker, Y. Luo,
M. Bewley, C. A. Smith, M. E. Murphy, Y. Wang, A. B. Mason, R. C.
Woodworth, G. D. Brayer, and E. N. Baker. Two high-resolution crystal
structures of the recombinant N-lobe of human transferrin reveal a stmctural
change implicated in iron release. Biochemistry, 37:7919-7928 (1998).
34. H. M. Schulman, J. Martinez-Medellin, and R. Sidloi. The reticulocyte-
mediated release of iron and bicarbonate from transferrin: effect of metabolic
inhibitors, Biochim. Biophys. Acta, 343:529-534 (1974).
35. A. Egyed. The significance of transferrin-bound bicarbonate in the uptake of
iron by reticulocytes, Biochim. Biophys. Acta, 304:805-813 (1973).
36. H. Sun, H. Li, and P. J. Sadler. Transferrin as a metal ion mediator, Chem.
Rev., 99 :2817-2842 (1999).
220
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
37. H. Sun, M. C. Cox, H. Li, A. B. Mason, R. C. Woodworth, and P, J. Sadler.
[1H,13C] NMR determination of the order of lobe loading of human
transferrin with iron: comparison with other metal ions, FEBS Lett., 422:315-
320 (1998).
38. H. Li, P. J. Sadler, and H. Sun. Rationalization of the strength of metal
binding to human serum transferrin, Eur. J. Biochem., 242:387-393 (1996).
39. M. Guo, H. Sun, H. J. McArdle, L. Gambling, and P. J. Sadler. Ti(IV) uptake
and release by human semm transferrin and recognition of Ti(IV)-transferrin
by cancer cells: understanding the mechanism of action of the anticancer drug
titanocene dichloride. Biochemistry, 39:10023-10033 (2000).
40. P. H. Jarritt. Effect of iron on sedimentation-velocity and gel filtration
behaviour of transferrins from several vertebrates, Biochim. Biophys. Acta,
453:332-343 (1976).
41. P. A. Charlwood. Differential sedimentation-velocity and gel-filtration
measurements on human apotransferrin and iron-transferrin, Biochem. J.,
125:1019-1026(1971).
42. D. G. Makey and U. S. Seal. The detection of four molecular forms of human
transferrin during the iron binding process, Biochim. Biophys. Acta, 453:250-
256 (1976).
43. H. A. Huebers, E. Csiba, E. Huebers, and C. A. Finch. Competitive
advantage of diferric transferrin in delivering iron to reticulocytes, Proc.
Natl. Acad. Sci. U . S. A, 80:300-304 (1983).
44. S. P. Young, A. Bomford, and R. Williams. The effect of the iron saturation
of transferrin on its binding and uptake by rabbit reticulocytes, Biochem. J.,
219:505-510 (1984).
45. R. M. Palmour and H. E. Sutton. Vertebrae transferrins. Molecular weights,
chemical compositions, and iron-binding studies. Biochemistry, 10:4026-
4032 (1971).
221
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
46. Y. Yeh, S. Iwai, and R. E. Feeney. Conformations of denatured and renatured
ovotransferrin, Biochemistry, 18:882-889 (1979).
47. J. G. Grossmann, A. B. Mason, R. C. Woodworth, M. Neu, P. F. Lindley, and
S. S. Hasnain. Asp ligand provides the trigger for closure of transferrin
molecules. Direct evidence from X-ray scattering studies of site-specific
mutants of the N-terminal half-molecule of human transferrin, J. Mol. Biol,
231:554-558 (1993).
48. Q. Y. He, A. B. Mason, B. M. Tam, R. T. MacGillivray, and R. C.
Woodworth. Dual role of Lys206-Lys296 interaction in human transferrin N-
lobe: iron-release trigger and anion-binding site. Biochemistry, 38:9704-9711
(1999).
49. P. Aisen. Transferrin metabolism and the liver, Semin. Liver Dis., 4:193-206
(1984).
50. Alper, C. A., Raum D., Awdeh Z.L., Petersen B.H., Taylor P.D., and Starzl
T.E. Studies of hepatic synthesis in vivo of plasma proteins, including
orosomucoid, transferrin, antitrypsin, C8, and factor B 1.
Chn.Immunol.Immunopathol. 16, 84-89. 11-11-1974.
Ref Type: Journal (Full)
51. E. H. Morgan and T. Peters, Jr. Intracellular aspects of transferrin synthesis
and secretion in the rat, J. Biol. Chem., 246:3508-3511 (1971).
52. E. H. Morgan and T. Peters, Jr. The biosynthesis of rat serum albumin. V.
Effect of protein depletion and refeeding on albumin and transferrin
synthesis, J. Biol Chem., 246:3500-3507 (1971).
53. A. G. Morton and A. S. Tavill. The role of iron in the regulation of hepatic
transferrin synthesis, Br. J. Haematol., 36:383-394 (1977).
54. Katz J.H. Iron and protein kinetic studies by means of a doubly labeled
human crystalline transferrin., J. Clin. Invest., 40:2143-2152 (1961).
222
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
55. I. S. Trowbridge and F. Lopez. Monoclonal antibody to transferrin receptor
blocks transferrin binding and inhibits human tumor cell growth in vitro,
Proc. Natl. Acad. Sci. U. S. A, 79:1175-1179 (1982).
56. B. Nikinmaa, C. A. Enns, S. E. Tonik, H. H. Sussman, and J. Schroder.
Monoclonal antibodies to a purified human transferrin receptor, Scand. J.
Immunol, 20:441-447 (1984).
57. J. W. Coding and G. F. Burns. Monoclonal antibody OKT-9 recognizes the
receptor for transferrin on human acute lymphocytic leukemia cells, J.
Immunol, 127:1256-1258 (1981).
58. T. A. Hamilton, H. G. Wada, and H. H. Sussman. Identification of transferrin
receptors on the surface of human cultured cells, Proc. Natl. Acad. Sci. U. S.
A, 76:6406-6410 (1979).
59. C. M. Lawrence, S . Ray, M. Babyonyshev, R. Galluser, D. W. Borhani, and
S. C. Harrison. Crystal structure of the ectodomain of human transferrin
receptor. Science, 286:779-782 (1999).
60. M. B. Omary and I. S. Trowbridge. Covalent binding of fatty acid to the
transferrin receptor in cultured human cells, J. Biol Chem., 256:4715-4718
(1981).
61. S. Q. Jing and I. S. Trowbridge. Identification of the intermolecular disulfide
bonds of the human transferrin receptor and its lipid-attachment site, EMBO
6:327-331 (1987).
62. A. M. Williams and C. A. Enns. A mutated transferrin receptor lacking
asparagine-linked glycosylation sites shows reduced functionality and an
association with binding immunoglobulin protein, J. Biol. Chem., 266:17648-
17654(1991).
63. E. A. Rutledge and C. A. Enns. Cleavage of the transferrin receptor is
influenced by the composition of the 0-linked carbohydrate at position 104,
J. Cell Physiol, 168:284-293 (1996).
223
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
64. I. S. Trowbridge, R. A. Newman, D. L. Domingo, and C. Sauvage.
Transferrin receptors: structure and function, Biochem. Pharmacol, 33:925-
932 (1984).
65. O. Zak, D. Trinder, and P. Aisen. Primary receptor-recognition site of human
transferrin is in the C- terminal lobe, J. Biol Chem., 269:7110-7114 (1994).
66. A. B. Mason, B. M. Tam, R. C. Woodworth, R. W. Oliver, B. N. Green, L. N.
Lin, J. F. Brandts, K. J. Savage, J. A. Lineback, and R. T. MacGillivray.
Receptor recognition sites reside in both lobes of human serum transferrin,
Biochem. J., 326 ( Ft l):77-85 (1997).
67. V. Dubljevic, A. Sali, and J. W. Goding. A conserved RGD (Arg-Gly-Asp)
motif in the transferrin receptor is required for binding to transferrin,
Biochem. J., 341 ( Ft 1):11-14 (1999).
68. R. D. Klausner, J. Van Renswoude, G. Ashwell, C. Kempf, A. N. Schechter,
A. Dean, and K. R. Bridges. Receptor-mediated endocytosis of transferrin in
K562 cells, J. Biol Chem., 258:4715-4724 (1983).
69. R. D. Klausner, G. Ashwell, J. Van Renswoude, J. B. Harford, and K. R.
Bridges. Binding of apotransferrin to K562 cells: explanation of the
transferrin cycle, Proc. Natl Acad. Sci U. S. A, 80:2263-2266 (1983).
70. J. Harford, K. Bridges, G. Ashwell, and R. D. Klausner. Intracellular
dissociation of receptor-bound asialoglycoproteins in cultured hepatocjdes. A
pH-mediated nonlysosomal event, J. Biol Chem., 258:3191-3197 (1983).
71. H. Kawabata, R. Yang, T. Hirama, P. T. Vuong, S. Kawano, A. F. Gombart,
and H. P. Koeffler. Molecular cloning of transferrin receptor 2. A new
member of the transferrin receptor-like family, J. Biol Chem. ,20826-20832
(1999).
72. R. E. Fleming, M. C. Migas, C. C. Holden, A. Waheed, R. S. Britton, S.
Tomatsu, B. R. Bacon, and W. S. Sly. Transferrin receptor 2: continued
expression in mouse liver in the face of iron overload and in hereditary
hemochromatosis, Proc. Natl Acad. Sci U. S. A, 97:2214-2219 (2000).
224
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
73. H. Kawabata, R. S. Germain, P. T. Vuong, T. Nakamaki, J. W. Said, and H.
P. Koeffler. Transferrin receptor 2-alpha supports cell growth both in iron-
chelated cultured cells and in vivo, J. Biol. Chem., 275:16618-16625 (2000).
74. H. Kawabata, R. S. Germain, T. Ikezoe, X. Tong, E. M. Green, A. F.
Gombart, and H. P. Koeffler. Regulation of expression of murine transferrin
receptor 2, Blood, 98 :1949-1954 (2001).
75. H. Kawabata, T. Nakamaki, P. Ikonomi, R. D. Smith, R. S. Germain, and H.
P. Koeffler. Expression of transferrin receptor 2 in normal and neoplastic
hematopoietic cells, Blood, 98:2714-2719 (2001).
76. M. De Gobbi, F. Daraio, C. Oberkanins, A. Moritz, F. Kury, G. Fiorelli, and
C. Camaschella. Analysis of HFE and TFR2 mutations in selected blood
donors with biochemical parameters of iron overload, Haematologica,
88:396-401 (2003).
77. A. Roetto, F. Daraio, F. Alberti, P. Porporato, A. Cali, M. De Gobbi, and C.
Camaschella. Hemochromatosis due to mutations in transferrin receptor 2,
Blood Cells Mol. Dis., 29:465-470 (2002).
78. A. Roetto, A. Totaro, A. Pipemo, A. Piga, F. Longo, G. Garozzo, A. Cali, M.
De Gobbi, P. Gasparini, and C. Camaschella. New mutations inactivating
transferrin receptor 2 in hemochromatosis type 3, Blood, 97:2555-2560
(2001).
79. C. Camaschella, A. Roetto, A. Cali, M. De Gobbi, G. Garozzo, M. Carella,
N. Majorano, A. Totaro, and P. Gasparini. The gene TFR2 is mutated in a
new type of haemochromatosis mapping to 7q22, Nat. Genet, 25:14-15
(2000).
80. R. E. Fleming and W. S. Sly. Hepcidin: a putative iron-regulatory hormone
relevant to hereditary hemochromatosis and the anemia of chronic disease.,
Proc. Natl. Acad. Sci. USA, 98:8160-8162 (2001).
225
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
81. J. L. Casey, M. W . Hentze, D. M. Koeller, S. W. Caughman, T. A. Rouault,
R. D. Klausner, and J. B. Harford. Iron-responsive elements: regulatory RNA
sequences that control mRNA levels and translation. Science, 240:924-928
(1988).
82. K. J. Addess, J. P. Basilion, R. D. Klausner, T. A. Rouault, and A. Pardi.
Structure and dynamics of the iron responsive element RNA: implications for
binding of the RNA by iron regulatory binding proteins, J. Mol. Biol,
274:72-83 (1997).
83. E. A. Leibold and H. N. Munro. Cytoplasmic protein binds in vitro to a
highly conserved sequence in the 5' untranslated region of ferritin heavy- and
light-subunit mRNAs, Proc. Natl Acad. Sci U. S. A, 85:2171-2175 (1988).
84. K. Rao, J. B. Harford, T. Rouault, A. McClelland, F. H. Ruddle, and R. D.
Klausner. Transcriptional regulation by iron of the gene for the transferrin
receptor. Mol. Cell Biol, 6:236-240 (1986).
85. E. W. Mullner and L. C. Kuhn. A stem-loop in the 3' untranslated region
mediates iron-dependent regulation of transferrin receptor mRNA stability in
the cytoplasm. Cell, 53:815-825 (1988).
86. R. Erlitzki, J. C. Long, and E. C. Theil. Multiple, conserved iron-responsive
elements in the 3'-untranslated region of transferrin receptor mRNA enhance
binding of iron regulatory protein 2, J. Biol. Chem., 277:42579-42587 (2002).
87. B. R. Henderson, C. Seiser, and L. C. Kuhn. Characterization of a second
RNA-binding protein in rodents with specificity for iron-responsive elements,
J. Biol Chem., 268:27327-27334 (1993).
88. C. Bouton, M. J. Chauveau, S. Lazereg, and J. C. Drapier. Recycling of RNA
binding iron regulatory protein 1 into an aconitase after nitric oxide removal
depends on mitochondrial ATP, J. Biol. Chem., 277:31220-31227 (2002).
89. J. Narahari, R. Ma, M. Wang, and W. E. Walden. The aconitase function of
iron regulatory protein 1. Genetic studies in yeast implicate its role in iron-
mediated redox regulation, J. Biol. Chem., 275:16227-16234 (2000).
226
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
90. A. Constable, S. Quick, N. K. Gray, and M. W. Hentze. Modulation of the
RNA-binding activity of a regulatory protein by iron in vitro: switching
between enzymatic and genetic function?, Proc. Natl. Acad. Sci. U. S. A,
89:4554-4558 (1992).
91. D. J. Haile, T. A. Rouault, C. K. Tang, J. Chin, J. B. Harford, and R. D.
Klausner. Reciprocal control of RNA-binding and aconitase activity in the
regulation of the iron-responsive element binding protein: role of the iron-
sulfur cluster, Proc. Natl. Acad. Sci. U. S. A , 89:7536-7540 (1992).
92. J. P. Basilion, M. C. Kennedy, H. Beinert, C. M. Massinople, R. D. Klausner,
and T. A. Rouault. Overexpression of iron-responsive element-binding
protein and its analytical characterization as the RNA-binding form, devoid
of an iron- sulfur cluster, Arch. Biochem. Biophys., 311:517-522 (1994).
93. J. Wang and K. Pantopoulos. Conditional derepression of ferritin synthesis in
cells expressing a constitutive IRPl mutant, Mol. Cell Biol, 22:4638-4651
(2002).
94. M. W. Hentze, S. W. Caughman, T. A. Rouault, J. G. Barriocanal, A. Dancis,
J. B. Harford, and R. D. Klausner. Identification of the iron-responsive
element for the translational regulation of human ferritin mRNA, Science,
238:1570-1573 (1987).
95. M. W. Hentze, T. A. Rouault, S. W. Caughman, A. Dancis, J. B. Harford, and
R. D. Klausner. A cis-acting element is necessary and sufficient for
translational regulation of human ferritin expression in response to iron,
Proc. Natl Acad. Sci U. S. A, 84:6730-6734 (1987).
96. C. R. Bhasker, G. Burgiel, B. Neupert, A. Emery-Goodman, L. C. Kuhn, and
B. K. May. The putative iron-responsive element in the human erythroid 5-
aminolevulinate synthase mRNA mediates translational control, J. Biol
Chem., 268:12699-12705 (1993).
227
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
97. K. Iwai, S. K. Drake, N. B. Wehr, A. M. Weissman, T. LaVaute, N. Minato,
R. D. Klausner, R. L. Levine, and T. A. Rouault. Iron-dependent oxidation,
ubiquitination, and degradation of iron regulatory protein 2: implications for
degradation of oxidized proteins, Proc. Natl. Acad. Sci. U. S. A, 95:4924-
4928 (1998).
98. K. Iwai, R. D. Klausner, and T. A. Rouault. Requirements for iron-regulated
degradation of the RNA binding protein, iron regulatory protein 2, EMBO J.,
14:5350-5357 (1995).
99. B. Guo, Y. Yu, and E. A. Leibold. Iron regulates cytoplasmic levels of a
novel iron-responsive element- binding protein without aconitase activity, J.
Biol. Chem., 269:24252-24260 (1994).
100. B. Guo, J. D. Phillips, Y. Yu, and E. A. Leibold. Iron regulates the
intracellular degradation of iron regulatory protein 2 by the proteasome, J.
Biol. C/zem., 270:21645-21651 (1995).
101. B. Guo, F. M. Brown, J. D. Phillips, Y. Yu, and E. A. Leibold.
Characterization and expression of iron regulatory protein 2 (IRP2). Presence
of multiple IRP2 transcripts regulated by intracellular iron levels, J. Biol.
Chem., 270:16529-16535 (1995).
102. B. R. Henderson and L. C. Kuhn. Differential modulation of the RNA-
binding proteins IRP-1 and IRP-2 in response to iron. IRP-2 inactivation
requires translation of another protein, J. Biol. Chem., 270:20509-20515
(1995).
103. P. A. DeRusso, C. C. Philpott, K. Iwai, H. S. Mostowski, R. D. Klausner, and
T. A. Rouault. Expression of a constitutive mutant of iron regulatory protein
1 abolishes iron homeostasis in mammalian cells, J. Biol. Chem., 270:15451-
15454(1995).
104. T. Rouault and R. Klausner. Regulation of iron metabolism in eukaryotes,
Curr. Top. Cell ReguL, 35:1-19 (1997).
228
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
105. X. Tong, H. Kawabata, and H. P. Koeffler. Iron deficiency can upregulate
expression of transferrin receptor at both the mRNA and protein level, Br. J.
Haematol, 116:458-464 (2002).
106. M. Davies, J. E. Parry, and R. G. Sutcliffe. Examination of different
preparations of human placental plasma membrane for the binding of insulin,
transferrin and immunoglobulins, J. Reprod. Fertil, 63:315-324 (1981).
107. C. A. Enns, J. E. Shindelman, S. E. Tonik, and H. H. Sussman.
Radioimmunochemical measurement of the transferrin receptor in human
trophoblast and reticulocyte membranes with a specific anti-receptor
antibody, Proc. Natl Acad. Sci U. S. A, 78:4222-4225 (1981).
108. K. C. Gatter, G. Brown, I. S. Trowbridge, R. E. Woolston, and D. Y. Mason.
Transferrin receptors in human tissues: their distribution and possible clinical
relevance, J. Clin. Pathol, 36:539-545 (1983).
109. M. Steiner. Fluorescence microphotometric studies of the transferrin receptor
in human erythroid precursor cells, J. Lab Clin. Med., 96:1086-1093 (1980).
110. H. G. Wada, P. E. Hass, and H. H. Sussman. Transferrin receptor in human
placental brush border membranes. Studies on the binding of transferrin to
placental membrane vesicles and the identification of a placental brush
border glycoprotein with high affinity for transferrin, J. Biol. Chem.,
254:12629-12635(1979).
111. W. A. Jefferies, M. R. Brandon, S. V. Hunt, A. F. Williams, K. C. Gatter, and
D. Y. Mason. Transferrin receptor on endothelium of brain capillaries,
Nature, 312:162-163 (1984).
112. D. Shah and W. C. Shen. Transcellular delivery of an insulin-transferrin
conjugate in enterocyte-like Caco-2 cells, J. Pharm. Sci, 85:1306-1311
(1996).
113. C. Q. Xia, J. Wang, and W. C. Shen. Hypoglycemic effect of insulin-
transferrin conjugate in streptozotocin- induced diabetic rats, J. Pharmacol.
Exp. Ther., 295:594-600 (2000).
229
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
114. Widera, A. W., Beloussow K., Kim K.J., Crandall E.D., and Shen W.C.
Phenotype-dependent synthesis of transferrin receptor in rat alveolar
epithelial cell monolayers. Cell & Tissue Research in press.
115. W. P. Faulk, B. L. Hsi, and P. J. Stevens. Transferrin and transferrin
receptors in carcinoma of the breast. Lancet, 2:390-392 (1980).
116. N. C. Andrews. Disorders of iron metabolism, N. Engl. J. Med., 341:1986-
1995 (1999).
117. J. Fuchs, M. Podda, L. Packer, and R. Kaufmann. Morbidity risk in HFE
associated hereditary hemochromatosis C282Y heterozygotes, Toxicology,
180:169-181 (2002).
118. J. N. Feder, A. Gnirke, W. Thomas, Z. Tsuchihashi, D. A. Ruddy, A. Basava,
F. Dormishian, R. Domingo, Jr., M. C. Ellis, A. Fullan, L. M. Hinton, N. L.
Jones, B. E. Kimmel, G. S. Eironmal, P. Lauer, V. K. Lee, D. B. Loeb, F. A.
Mapa, E. McClelland, N. C. Meyer, G. A. Mintier, N. Moeller, T. Moore, E.
Morikang, R. K. Wolff, and. A novel MHC class I-like gene is mutated in
patients with hereditary haemochromatosis, Nat. Genet., 13:399-408 (1996).
119. J. M. Bastin, M. Jones, C. A. O'Callaghan, L. Schimanski, D. Y. Mason, and
A. R. Townsend. Kupffer cell staining by an HFE-specific monoclonal
antibody: implications for hereditary haemochromatosis, Br. J. Haematol,
103:931-941 (1998).
120. S. Parkkila, A. Waheed, R. S. Britton, J. N. Feder, Z. Tsuchihashi, R. C.
Schatzman, B. R. Bacon, and W. S. Sly. Immunohistochemistry of HLA-H,
the protein defective in patients with hereditary hemochromatosis, reveals
unique pattern of expression in gastrointestinal tract, Proc. Natl. Acad. Sci. U .
S. A, 94:2534-2539 (1997).
121. J. A. Lebron, M. J. Bennett, D. E. Vaughn, A. J. Chirino, P. M. Snow, G. A.
Mintier, J. N. Feder, and P. J. Bjorkman. Crystal structure of the
hemochromatosis protein HFE and characterization of its interaction with
transferrin receptor. Cell, 93:111-123 (1998).
230
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
122. M. J. Bennett, J. A. Lebron, and P. J. Bjorkman. Crystal structure of the
hereditary haemochromatosis protein HFE complexed with transferrin
receptor. Nature, 403:46-53 (2000).
123. J. N. Feder, D. M. Penny, A. Irrinki, V. K. Lee, J. A. Lebron, N. Watson, Z.
Tsuchihashi, E. Sigal, P. J. Bjorkman, and R. C. Schatzman. The
hemochromatosis gene product complexes with the transferrin receptor and
lowers its affinity for ligand binding, Proc. Natl. Acad. Sci. U. S. A, 95:1472-
1477 (1998).
124. J. N. Feder, Z. Tsuchihashi, A. Irrinki, V. K. Lee, F. A. Mapa, E. Morikang,
C. E. Prass, S. M. Starnes, R. K. Wolff, S. Parkkila, W. S. Sly, and R. C.
Schatzman. The hemochromatosis founder mutation in HLA-H disrupts
beta2- microglobulin interaction and cell surface expression, J. Biol. Chem.,
272:14025-14028(1997).
125. A. Waheed, S. Parkkila, X. Y. Zhou, S. Tomatsu, Z. Tsuchihashi, J. N. Feder,
R. C. Schatzman, R. S. Britton, B. R. Bacon, and W. S. Sly. Hereditary
hemochromatosis: effects of C282Y and H63D mutations on association with
beta2-microglobulin, intracellular processing, and cell surface expression of
the HFE protein in COS-7 cells, Proc. Natl. Acad. Sci. U . S. A, 94:12384-
12389 (1997).
126. S. Parkkila, A. Waheed, R. S. Britton, B. R. Bacon, X. Y. Zhou, S. Tomatsu,
R. E. Fleming, and W. S. Sly. Association of the transferrin receptor in
human placenta with HFE, the protein defective in hereditary
hemochromatosis, Proc. Natl. Acad. Sci. U. S. A, 94:13198-13202 (1997).
127. C. N. Gross, A. Irrinki, J. N. Feder, and C. A. Enns. Co-trafficking of HFE, a
nonclassical major histocompatibility complex class I protein, with the
transferrin receptor implies a role in intracellular iron regulation, J. Biol.
Chem., 273:22068-22074 (1998).
128. X. Y. Zhou, S. Tomatsu, R. E. Fleming, S. Parkkila, A. Waheed, J. Jiang, Y.
Fei, E. M. Brunt, D. A. Ruddy, C. E. Prass, R. C. Schatzman, R. O'Neill, R.
S. Britton, B. R. Bacon, and W. S. Sly. HFE gene knockout produces mouse
model of hereditary hemochromatosis, Proc. Natl. Acad. Sci. U . S. A,
95:2492-2497 (1998).
231
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
129. J. A. Lebron, A. P. West, Jr., and P. J. Bjorkman. The hemochromatosis
protein HFE competes with transferrin for binding to the transferrin receptor,
J Mol. Biol., 294:239-245 (1999).
130. C. N. Roy, D. M. Penny, J. N. Feder, and C. A. Enns. The hereditary
hemochromatosis protein, HFE, specifically regulates transferrin-mediated
iron uptake in HeLa cells, J. Biol. Chem., 274:9022-9028 (1999).
131. H. D. Riedel, M. U. Muckenthaler, S. G. Gehrke, I. Mohr, K. Brennan, T.
Herrmann, B. A. Fitscher, M. W. Hentze, and W. Stremmel. HFE
downregulates iron uptake irom transferrin and induces iron- regulatory
protein activity in stably transfected cells. Blood, 94:3915-3921 (1999).
132. B. Corsi, S. Levi, A. Cozzi, A. Corti, D. Altimare, A. Albertini, and P.
Arosio. Overexpression of the hereditary hemochromatosis protein, HFE, in
HeLa cells induces and iron-deficient phenotype, FEBS Lett.., 460:149-152
(1999).
133. T. S. Ramalingam, A. P. West, Jr., J. A. Lebron, J. S. Nangiana, T. H. Hogan,
C. A. Enns, and P. J. Bjorkman. Binding to the transferrin receptor is required
for endocytosis of HFE and regulation of iron homeostasis, Nat. Cell Biol,
2:953-957 (2000).
134. A. P. West, Jr., M. J. Bennett, V. M. Sellers, N. C. Andrews, C. A. Enns, and
P. J. Bjorkman. Comparison of the interactions of transferrin receptor and
transferrin receptor 2 with transferrin and the hereditary hemochromatosis
protein HFE, J. Biol Chem., 275:38135-38138 (2000).
135. J. E. Levy, L. K. Montross, D. E. Cohen, M. D. Fleming, and N. C. Andrews.
The C282Y mutation causing hereditary hemochromatosis does not produce a
null allele. Blood, 94:9-11 (1999).
136. B. Seetharam, E. I. Christensen, S. K. Moestrup, T. G. Hammond, and P. J.
Verroust. Identification of rat yolk sac target protein of teratogenic
antibodies, gp280, as intrinsic factor-cobalamin receptor, J. Clin. Invest,
99:2317-2322(1997).
232
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
137. S. K. Moestrup, R. Kozyraki, M. Kristiansen, J. H. Kaysen, H. H.
Rasmussen, D. Brault, F. Pontillon, F. O. Goda, E. I. Christensen, T. G.
Hammond, and P. J. Verroust. The intrinsic factor-vitamin B12 receptor and
target of teratogenic antibodies is a megalin-binding peripheral membrane
protein with homology to developmental proteins, J. Biol. Chem., 273:5235-
5242 (1998).
138. R. Kozyraki, M. Kristiansen, A. Silahtaroglu, C. Hansen, C. Jacobsen, N.
Tommerup, P. J. Verroust, and S. K. Moestrup. The human intrinsic factor-
vitamin B12 receptor, cubilin: molecular characterization and chromosomal
mapping of the gene to lOp within the autosomal recessive megaloblastic
anemia (MGAl) region. Blood, 9 1 :3593-3600 (1998).
139. D. Xu, R. Kozyraki, T. C. Newman, and J. C. Fyfe. Genetic evidence of an
accessory activity required specifically for cubilin brush-border expression
and intrinsic factor-cobalamin absorption. Blood, 94:3604-3606 (1999).
140. P. Bork and G. Beckmann. The CUB domain. A widespread module in
developmentally regulated proteins, J. Mol. Biol, 231:539-545 (1993).
141. M. Kristiansen, R. Kozyraki, C. Jacobsen, E. Nexo, P. J. Verroust, and S. K.
Moestrup. Molecular dissection of the intrinsic factor-vitamin B12 receptor,
cubilin, discloses regions important for membrane association and ligand
binding,/. Biol Chem., 274:20540-20544 (1999).
142. V. K. Mishra and M. N. Palgunachari. Interaction of model class A l, class
A2, and class Y amphipathic helical peptides with membranes. Biochemistry,
35:11210-11220 (1996).
143. C. Mercier, M. F. Cesbron-Delauw, and L. D. Sibley. The amphipathic alpha
helices of the toxoplasma protein GRA2 mediate post-secretory membrane
association, J. Cell Sci, 111 ( Pt 15):2171-2180 (1998).
144. Y. Li, T. Smith, S. Grabski, and D. L. DeWitt. The membrane association
sequences of the prostaglandin endoperoxide synthases-1 and -2 isozymes, J.
Biol Chem., 273:29830-29837 (1998).
233
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
145. C. Chen, K. T. Seow, K. Guo, L. P. Yaw, and S. C. Lin. The membrane
association domain of RGS16 contains unique amphipathic features that are
conserved in RGS4 and RGS5, J. Biol. Chem., 274:19799-19806 (1999).
146. L. S. Bernstein, A. A. Grillo, S. S. Loranger, and M. E. Linder. RGS4 binds
to membranes through an amphipathic alpha -helix, J. Biol. Chem.,
275:18520-18526(2000).
147. K. S. Ramanujam, S. Seetharam, N. M. Dahms, and B. Seetharam. Effect of
processing inhibitors on cobalamin (vitamin B12) transcytosis in polarized
opossum kidney cells. Arch. Biochem. Biophys., 315:8-15 (1994).
148. S. K. Moestrup and P. J. Verroust. Megalin- and cubilin-mediated
endocytosis of protein-bound vitamins, lipids, and hormones in polarized
epithelia, Annu. Rev. Nutr., 21:407-428 (2001).
149. R. Raychowdhury, J. L. Niles, R. T. McCluskey, and J. A. Smith.
Autoimmune target in Heymann nephritis is a glycoprotein with homology to
the LDL receptor. Science, 244:1163-1165 (1989).
150. A. Saito, S. Pietromonaco, A. K. Loo, and M. G. Farquhar. Complete cloning
and sequencing of rat gp330/"megalin," a distinctive member of the low
density lipoprotein receptor gene family, Proc. Natl. Acad. Sci. U. S. A,
91:9725-9729(1994).
151. G. Hjalm, E. Murray, G. Cmmley, W. Harazim, S. Lundgren, I. Onyango, B.
Ek, M. Larsson, C. Juhlin, P. Heilman, H. Davis, G. Akerstrom, L. Rask, and
B. Morse. Cloning and sequencing of human gp330, a Ca(2+)-binding
receptor with potential intracellular signaling properties, Eur. J. Biochem.,
239:132-137(1996).
152. C. G. Davis, J. L. Goldstein, T. C. Sudhof, R. G. Anderson, D. W. Russell,
and M. S. Brown. Acid-dependent ligand dissociation and recycling of LDL
receptor mediated by growth factor homology region. Nature, 326:760-765
(1987).
234
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
153. W. J. Chen, J. L. Goldstein, and M. S. Brown. NPXY, a sequence often found
in cytoplasmic tails, is required for coated pit-mediated internalization of the
low density lipoprotein receptor, J. Biol. Chem., 265:3116-3123 (1990).
154. T. E. Willnow, A. Nykjaer, and J. Herz. Lipoprotein receptors: new roles for
ancient proteins, Nat. Cell Biol, 1:E 157-El62 (1999).
155. C. Juhlin, S. Lundgren, H. Johansson, J. Lorentzen, L. Rask, E. Larsson, J.
Rastad, G. Akerstrom, and L. Klareskog. 500-Kilodalton calcium sensor
regulating cytoplasmic Ca2+ in cytotrophoblast cells of human placenta, J.
Biol Chem., 265:8275-8279 (1990).
156. D. Sahali, N. Mulliez, F. Chatelet, C. Laurent-Winter, D. Citadelle, C. Roux,
P. Ronco, and P. Verroust. Coexpression in humans by kidney and fetal
envelopes of a 280 kDa- coated pit-restricted protein. Similarity with the
murine target of teratogenic antibodies, Am. J. Pathol, 140:33-44 (1992).
157. D. Sahali, N. Mulliez, F. Chatelet, R. Dupuis, P. Ronco, and P. Verroust.
Characterization of a 280-kD protein restricted to the coated pits of the renal
brush border and the epithelial cells of the yolk sac. Teratogenic effect of the
specific monoclonal antibodies, J. Exp. Med., 167:213-218 (1988).
158. F. Chatelet, E. Brianti, P. Ronco, J. Roland, and P. Verroust. Ultrastructural
localization by monoclonal antibodies of brush border antigens expressed by
glomeruli. U. Extrarenal distribution. Am. J. Pathol, 122:512-519 (1986).
159. D. Kerjaschki and M. G. Farquhar. The pathogenic antigen of Heymann
nephritis is a membrane glycoprotein of the renal proximal tubule brush
border, Proc. Natl Acad. Scl U. S. A, 79:5557-5581 (1982).
160. B. Seetharam, J. S. Levine, M. Ramasamy, and D. H. Alpers. Purification,
properties, and immunochemical localization of a receptor for intrinsic
factor-cobalamin complex in the rat kidney, J. Biol Chem., 263:4443-4449
(1988).
235
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
161. F. Chatelet, E. Brianti, P. Ronco, J. Roland, and P. Verroust. Ultrastructural
localization by monoclonal antibodies of brush border antigens expressed by
glomeruli. I. Renal distribution. Am. J. Pathol, 122:500-511 (1986).
162. J. S. Levine, R. H. Allen, D. H. Alpers, and B. Seetharam.
Immunocytochemical localization of the intrinsic factor-cobalamin receptor
in dog-ileum: distribution of intracellular receptor during cell maturation, J.
Cell Biol ,98:1111-1118(1984).
163. H. Bim, P. J. Verroust, E. Nexo, H. Hager, C. Jacobsen, E. I. Christensen,
and S. K. Moestrup. Characterization of an epithelial approximately 460-kDa
protein that facilitates endocytosis of intrinsic factor-vitamin B12 and binds
receptor-associated protein, J. Biol Chem., 272:26497-26504 (1997).
164. D. Kerjaschki, L. Noronha-Blob, B. Sacktor, and M. G. Farquhar.
Microdomains of distinctive glycoprotein composition in the kidney proximal
tubule brush border, J. Cell B iol, 98:1505-1513 (1984).
165. E. I. Christensen, J. Gliemann, and S. K. Moestrup. Renal tubule gp330 is a
calcium binding receptor for endocytic uptake of protein, J. Histochem.
Cytochem., 40:1481-1490 (1992).
166. R. Kozyraki, J. Fyfe, P. J. Verroust, C. Jacobsen, A. Dautry-Varsat, J.
Gburek, T. E. Willnow, E. I. Christensen, and S. K. Moestrup. Megalin-
dependent cubilin-mediated endocytosis is a major pathway for the apical
uptake of transferrin in polarized epithelia, Proc. Natl. Acad. Scl U. S. A,
98:12491-12496(2001).
167. J. E. Casanova, X. Wang, R. Kumar, S. G. Bhartur, J. Navarre, J. E.
Woodrum, Y. Altschuler, G. S. Ray, and J. R. Goldenring. Association of
Rab25 and Rabl la with the apical recycling system of polarized Madin-
Darby canine kidney cells. M ol Biol Ce//,47-61 (1999).
168. R. G. Parton. Endocytosis in polarized cells, Semin. Cell B iol, 2:387-395
(1991).
236
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
169. K. E. Mostov and M. H. Cardone. Regulation of protein traffic in polarized
epithelial cells, Bioessays, 17:129-138 (1995).
170. E. Wang, P. S. Brown, B. Aroeti, S. J. Chapin, K. E. Mostov, and K. W.
Dunn. Apical and basolateral endocytic pathways of MDCK cells meet in
acidic common endosomes distinct from a nearly-neutral apical recycling
endosome. Traffic., 1:480-493 (2000).
171. C. E. Putter, A. Gibson, E. H. Allchin, S. Maxwell, L. J. Ruddock, G.
Odorizzi, D. Domingo, I. S. Trowbridge, and C. R. Hopkins. In polarized
MDCK cells basolateral vesicles arise from clathrin-gamma- adaptin-coated
domains on endosomal tubules, J. Cell B iol, 141:611-623 (1998).
172. G. Odorizzi, A. Pearse, D. Domingo, I. S. Trowbridge, and C. R. Hopkins.
Apical and basolateral endosomes of MDCK cells are interconnected and
contain a polarized sorting mechanism, J. Cell B iol, 135:139-152 (1996).
173. B. Buendia, M. H. Bre, G. Griffiths, and E. Karsenti. Cytoskeletal control of
centrioles movement during the establishment of polarity in Madin-Darby
canine kidney cells, J. Cell B iol, 110:1123-1135 (1990).
174. D. R. Sheff, E. A. Daro, M. Hull, and I. Mellman. The receptor recycling
pathway contains two distinct populations of early endosomes with different
sorting fimctions, J. Cell B iol, 145:123-139 (1999).
175. D. Sheff, L. Pelletier, C. B. O'Connell, G. Warren, and I. Mellman.
Transferrin receptor recycling in the absence of perinuclear recycling
endosomes, J. Cell B iol, 156:797-804 (2002).
176. Y. Takai, T. Sasaki, and T. Matozaki. Small GTP-binding proteins, Physiol
i?ev., 81:153-208 (2001).
177. B. Sonnichsen, S. De Renzis, E. Nielsen, J. Rietdorf, and M. Zerial. Distinct
membrane domains on endosomes in the recycling pathway visualized by
multicolor imaging of Rab4, Rab5, and Rabl 1, J. Cell B iol, 149:901-914
(2000).
237
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
178. R. Gagescu, N. Demaurex, R. G. Parton, W. Hunziker, L. A. Huber, and J.
Gmenberg. The recycling endosome of Madin-Darby canine kidney cells is a
mildly acidic compartment rich in raft components. Mol. Biol. Cell, 11:2775-
2791 (2000).
179. M. Trischler, W. Stoorvogel, and O. Ullrich. Biochemical analysis of distinct
Rab5- and Rabl 1-positive endosomes along the transferrin pathway, J. Cell
Scl, 112 ( Pt 24):4773-4783 (1999).
180. O. Ullrich, S. Reinsch, S. Urbe, M. Zerial, and R. G. Parton. Rabl 1 regulates
recycling through the pericentriolar recycling endosome, J. Cell Biol,
135:913-924 (1996).
181. B. P. Ceresa, M. Lotscher, and S. L. Schmid. Receptor and membrane
recycling can occur with unaltered efficiency despite dramatic Rab5(q791)-
induced changes in endosome geometry, J. Biol Chem., 276:9649-9654
(2001).
182. C. Bucci, A. Lutcke, O. Steele-Mortimer, V. M. Olkkonen, P. Dupree, M.
Chiariello, C. B. Bruni, K. Simons, and M. Zerial. Co-operative regulation of
endocytosis by three Rab5 isoforms, FEBSLett., 366:65-71 (1995).
183. D. B. Wilson and M. P. Wilson. Identification and subcellular localization of
human rab5b, a new member of the ras-related superfamily of GTPases, J.
Clin. Invest, 89:996-1005 (1992).
184. M. Chiariello, C. B. Bruni, and C. Bucci. The small GTPases Rab5a, Rab5b
and Rab5c are differentially phosphorylated in vitro, FEBS Lett., 453:20-24
(1999).
185. E. Daro, S. P. van der, T. Galli, and I. Mellman. Rab4 and cellubrevin define
different early endosome populations on the pathway of transferrin receptor
recycling, Proc. Natl. Acad. Scl U. S. A,9559-9564 (1996).
186. G. Bottger, B. Nagelkerken, and S. P. van der. Rab4 and Rab7 define distinct
nonoverlapping endosomal compartments, J. Biol. Chem., 271:29191-29197
(1996).
238
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
187. S. P. van der, M. Hull, L. A. Huber, P. Male, B. Goud, and I. Mellman.
Reversible phosphorylation— dephosphorylation determines the localization
of rab4 during the cell cycle, EMBOJ., 11:4379-4389 (1992).
188. K. Mohrmann, R. Leijendekker, L. Gerez, and S. P. van der. rab4 regulates
transport to the apical plasma membrane in Madin-Darby canine kidney cells,
J Biol. Chem., 277:10474-10481 (2002).
189. K. Mohrmann, L. Gerez, V. Oorschot, J. Klumperman, and S. P. van der.
Rab4 function in membrane recycling from early endosomes depends on a
membrane to cytoplasm cycle, J. Biol. Chem., 277:32029-32035 (2002).
190. P. Chavrier, S. P. van der, Z. Mishal, B. Nagelkerken, and J. P. Gorvel. Early
endosome membrane dynamics characterized by flow cytometry. Cytometry,
29:41-49 (1997).
191. A. Durrbach, D. Louvard, and E. Coudrier. Actin filaments facilitate two
steps of endocytosis, J. Cell Sci., 109 ( Pt 2):457-465 (1996).
192. D. R. Sheff, R. Kroschewski, and I. Mellman. Actin dependence of polarized
receptor recycling in Madin-Darby canine kidney cell endosomes. Mol. Biol.
Cell, 13:262-275 (2002).
193. A. Durrbach, G. Raposo, D. Tenza, D. Louvard, and E. Coudrier. Truncated
brush border myosin I affects membrane traffic in polarized epithelial cells,
Traffic.,A\lA2A (2000).
194. J. R. Goldenring, J. Smith, H. D. Vaughan, P. Cameron, W. Hawkins, and J.
Navarre. Rabl 1 is an apically located small GTP-binding protein in epithelial
tissues. Am J Physiol, 270:G515-G525 (1996).
195. E. G. Green, E. Ramm, N. M. Riley, D. J. Spiro, J. R. Goldenring, and M.
Wessling-Resnick. Rabl 1 is associated with transferrin-containing recycling
compartments in K562 cells, Biochem. Biophys. Res. Commun., 239:612-616
(1997).
239
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
196. M. Ren, G. Xu, J. Zeng, C. Lemos-Chiarandini, M. Adesnik, and D. D.
Sabatini. Hydrolysis of GTP on rabl 1 is required for the direct delivery of
transferrin from the pericentriolar recycling compartment to the cell surface
but not from sorting endosomes, Proc. Natl. Acad. Sci. U. S. A, 95:6187-6192
(1998).
197. S. Urbe, L. A. Huber, M. Zerial, S. A. Tooze, and R. G. Parton. R abll, a
small GTPase associated with both constitutive and regulated secretory
pathways in PCI 2 cells, FEBS Lett., 334:175-182 (1993).
198. W. Chen, Y. Feng, D. Chen, and A. Wandinger-Ness. Rabl 1 is required for
trans-golgi network-to-plasma membrane transport and a preferential target
for GDP dissociation inhibitor. Mol. Biol. Cell, 9:3241-3257 (1998).
199. B. Schlierf, G. H. Fey, J. Hauber, G. M. Hocke, and 0. Rosorius. Rabl lb is
essential for recycling of transferrin to the plasma membrane. Exp. Cell Res.,
259:257-265 (2000).
200. R. Prekeris, J. Klumperman, and R. H. Scheller. A Rabl 1/Ripll protein
complex regulates apical membrane trafficking via recycling endosomes,
Mol. Cell, 6:1437-1448 (2000).
201. A. J. Lindsay, A. G. Hendrick, G. Cantalupo, F. Senic-Matuglia, B. Goud, C.
Bucci, and M. W. McCaffrey. Rab coupling protein (RCP), a novel Rab4 and
R abll effector protein, J. Biol. Chem., 277:12190-12199 (2002).
202. C. M. Hales, R. Griner, K. C. Hobdy-Henderson, M. C. Dorn, D. Hardy, R.
Kumar, J. Navarre, E. K. Chan, L. A. Lapierre, and J. R. Goldenring.
Identification and characterization of a family of Rabl 1-interacting proteins,
J. Biol. Chem., 276:39067-39075 (2001).
203. D. M. Wallace, A. J. Lindsay, A. G. Hendrick, and M. W. McCaffrey. The
novel Rab 11 -FIP/Rip/RCP family of proteins displays extensive homo- and
hetero-interacting abilities, Biochem. Biophys. Res. Commun., 292:909-915
(2002).
240
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
204. A. J. Lindsay and M. W. McCaffrey. Rabl 1-FIP2 fimctions in transferrin
recycling and associates with endosomal membranes via its COOH-terminal
domain, J Biol. Chem., 277:27193-27199 (2002).
205. A. Simonsen, R. Lippe, S. Christoforidis, J. M. Gaullier, A. Brech, J.
Callaghan, B. H. Toh, C. Murphy, M. Zerial, and H. Stenmark. EEAl links
PI(3)K fimction to RabS regulation of endosome fusion. Nature, 394:494-498
(1998).
206. F. T. Mu, J. M. Callaghan, O. Steele-Mortimer, H. Stenmark, R. G. Parton, P.
L. Campbell, J. McCluskey, J. P. Yeo, E. P. Lock, and B. H. Toh. EEAl, an
early endosome-associated protein. EEAl is a conserved alpha- helical
peripheral membrane protein flanked by cysteine "fingers" and contains a
calmodulin-binding IQ motif, J. Biol. Chem., 270:13503-13511 (1995).
207. H. M. McBride, V. Rybin, C. Murphy, A. Giner, R. Teasdale, and M. Zerial.
Oligomeric complexes link Rab5 effectors with NSF and drive membrane
fusion via interactions between EEAl and syntaxin 13, Cell, 98:377-386
(1999).
208. S. Christoforidis, H. M. McBride, R. D. Burgoyne, and M. Zerial. The Rab5
effector EEAl is a core component of endosome docking, Nature, 397:621-
625 (1999).
209. J. E. Rothman and G. Warren. Implications of the SNARE hypothesis for
intracellular membrane topology and dynamics, Curr. Biol, 4:220-233
(1994).
210. A. Simonsen, J. M. Gaullier, A. D'Arrigo, and H. Stenmark. The Rab5
effector EEAl interacts directly with syntaxin-6, J. Biol. Chem., 274:28857-
28860 (1999).
211. D. C. Lawe, A. Chawla, E. Merithew, J. Dumas, W. Carrington, K. Fogarty,
L. Lifshitz, R. Tuft, D. Lambright, and S. Corvera. Sequential roles for
phosphatidylinositol 3-phosphate and Rab5 in tethering and fusion of early
endosomes via their interaction with EEAl, J. Biol. Chem., 277:8611-8617
(2002).
241
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
212. H. Stenmark, R. Aasland, B. H. Toh, and A. D'Arrigo. Endosomal
localization of the autoantigen EEAl is mediated by a zinc- binding FYVE
fmger, J. Biol. Chem., I l l :24048-24054 (1996).
213. J. M. Gaullier, A. Simonsen, A. D'Arrigo, B. Bremnes, H. Stenmark, and R.
Aasland. FYVE fingers bind PtdIns(3)P, Nature, 394:432-433 (1998).
214. V. Patki, D. C. Lawe, S. Corvera, J. V. Virbasius, and A. Chawla. A
functional PtdIns(3)P-binding motif. Nature, 394:433-434 (1998).
215. C. G. Burd and S. D. Emr. Phosphatidylinositol(3)-phosphate signaling
mediated by specific binding to RING FYVE domains, Mol. Cell, 2:157-162
(1998).
216. C. Wiedemann and S. Cockcroft. Vesicular transport. Sticky fingers grab a
lipid. Nature, 394:426-427 (1998).
217. D. J. Gillooly, A. Simonsen, and H. Stenmark. Cellular fimctions of
phosphatidylinositol 3-phosphate and FYVE domain proteins, Biochem. J.,
355:249-258 (2001).
218. S. Christoforidis, M. Miaczynska, K. Ashman, M. Wilm, L. Zhao, S. C. Yip,
M. D. Waterfield, J. M. Backer, and M. Zerial. Phosphatidylinositol-3-OH
kinases are RabS effectors, Nat. Cell B iol, 1:249-252 (1999).
219. U. Siddhanta, J. Mcllroy, A. Shah, Y. Zhang, and J. M. Backer. Distinct roles
for the pllOalpha and hVPS34 phosphatidylinositol 3'- kinases in vesicular
trafficking, regulation of the actin cytoskeleton, and mitogenesis, J. Cell
B iol, 143:1647-1659(1998).
220. R. J. Davis, S. Corvera, and M. P. Czech. Insulin stimulates cellular iron
uptake and causes the redistribution of intracellular transferrin receptors to
the plasma membrane, J. Biol Chem. , 261:8708-8711 (1986).
221. R. Meyers and L. C. Cantley. Cloning and characterization of a wortmannin-
sensitive human phosphatidylinositol 4-kinase, J. Biol. Chem., 272:4384-
4390 (1997).
242
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
222. C. J. Vlahos, W. F. Matter, K. Y. Hui, and R. F. Brown. A specific inhibitor
of phosphatidylinositol 3-kinase, 2-(4- morpholinyl)-8-phenyl-4H-1 -
benzopyran-4-one (LY294002),/. Biol CAem.,5241-5248 (1994).
223. W. J. Brown, D. B. DeWald, S. D. Emr, H. Plutner, and W. E. Balch. Role
for phosphatidylinositol 3-kinase in the sorting and transport of newly
synthesized lysosomal enzymes in mammalian cells, J. Cell Biol, 130:781-
796 (1995).
224. N. A. Bright, M. R. Lindsay, A. Stewart, and J. P. Luzio. The relationship
between lumenal and limiting membranes in swollen late endocytic
compartments formed after wortmannin treatment or sucrose accumulation.
Traffic.,631-6A2 (2001).
225. J. L. Martys, C. Wjasow, D. M. Gangi, M. C. Kielian, T. E. McGraw, and J.
M. Backer. Wortmannin-sensitive trafficking pathways in Chinese hamster
ovary cells. Differential effects on endocytosis and lysosomal sorting, J. Biol
Chem., 271:10953-10962 (1996).
226. H. Shpetner, M. Joly, D. Hartley, and S. Corvera. Potential sites of PI-3
kinase function in the endocytic pathway revealed by the PI-3 kinase
inhibitor, wortmannin, J. Cell Biol, 132:595-605 (1996).
227. L. Hunyady, A. J. Baukal, Z. Gaborik, J. A. Olivares-Reyes, M. Bor, M.
Szaszak, R. Lodge, K. J. Catt, and T. Balia. Differential PI 3-kinase
dependence of early and late phases of recycling of the internalized ATI
angiotensin receptor, J. Cell Biol, 157:1211-1222 (2002).
228. E. M. van Dam, T. Ten Broeke, K. Jansen, P. Spijkers, and W. Stoorvogel.
Endocytosed transferrin receptors recycle via distinct dynamin and
phosphatidylinositol 3-kinase-dependent pathways, J. Biol Chem.,
277:48876-48883 (2002).
229. E. M. van Dam and W. Stoorvogel. Dynamin-dependent transferrin receptor
recycling by endosome-derived clathrin-coated vesicles. M ol Biol Cell,
13:169-182 (2002).
243
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
230. J. M. Lloyd, T. O'Dowd, M. Driver, and D. E. Tee. Demonstration of an
epitope of the transferrin receptor in human cervical epithelium-a potentially
useful cell marker, J. Clin. Pathol, 37:131-135 (1984).
231. H. N. Raaf, D. W. Jacobsen, S. Savon, and R. Green. Serum transferrin
receptor level is not altered in invasive adenocarcinoma of the breast. Am. J.
Clin. Pathol, 99:232-237 (1993).
232. L. D. Recht, T. W. Griffin, V. Raso, and A. R. Salimi. Potent cytotoxicity of
an antihuman transferrin receptor-ricin A-chain immunotoxin on human
glioma cells in vitro, Cancer Res., 50:6696-6700 (1990).
233. A. C. Frost, F. Menegaux, P. Langlois, J. M. Vidal, M. Koulibaly, J. L. lost,
J. J. Duron, J. P. Chigot, P. Vayre, A. Aurengo, J. C. Legrand, G. Rosselin,
and C. Gespach. Differential transferrin receptor density in human colorectal
cancer: A potential probe for diagnosis and therapy. Int. J. Oncol, 13:871-
875 (1998).
234. Y. Beguin, S. Lampertz, D. De Groote, D. Igot, M. Malaise, and G. Fillet.
Soluble CD23 and other receptors (CD4, CD8, CD25, CD71) in serum of
patients with chronic lymphocytic leukemia. Leukemia, 7:2019-2025 (1993).
235. J. E. Shindelman, A. E. Ortmeyer, and H. H. Sussman. Demonstration of the
transferrin receptor in human breast cancer tissue. Potential marker for
identifying dividing cells. Int. J. Cancer, 27:329-334 (1981).
236. J. W. Larrick and P. Cresswell. Modulation of cell surface iron transferrin
receptors by cellular density and state of activation, J. Supramol Struct.,
11:579-586 (1979).
237. R. Sutherland, D. Delia, C. Schneider, R. Newman, J. Kemshead, and M.
Greaves. Ubiquitous cell-surface glycoprotein on tumor cells is proliferation-
associated receptor for transferrin, Proc. N atl Acad. Scl U S. A, 78:4515-
4519(1981).
244
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
238. F. Kratz, U. Beyer, T. Roth, N. Tarasova, P. Collery, F. Lechenault, A.
Cazabat, P. Schumacher, C. Unger, and U. Falken. Transferrin conjugates of
doxorubicin: synthesis, characterization, cellular uptake, and in vitro efficacy,
J Pharm. Sci., 87:338-346 (1998).
239. F. Kratz, T. Roth, I. Fichiner, P. Schumacher, H. H. Fiebig, and C. Unger. In
vitro and in vivo efficacy of acid-sensitive transferrin and albumin
doxorubicin conjugates in a human xenograft panel and in the MDA-MB-435
mamma carcinoma model, J. Drug Target, 8:305-318 (2000).
240. M. Singh. Transferrin As A targeting ligand for liposomes and anticancer
drugs, Curr. Pharm. Des, 5:443-451 (1999).
241. J. Huwyler, J. Yang, and W. M. Pardridge. Receptor mediated delivery of
daunomycin using immunoliposomes: pharmacokinetics and tissue
distribution in the rat, J. Pharmacol. Exp. Ther., 282:1541-1546 (1997).
242. E. Wagner, M. Gotten, R. Foisner, and M. L. Bimstiel. Transferrin-
polycation-DNA complexes: the effect of polycations on the structure of the
complex and DNA delivery to cells, Proc. Natl. Acad. Sci. U. S. A, 88:4255-
4259(1991).
243. M. Gotten, F. Langle-Rouault, H. Kirlappos, E. Wagner, K. Mechtler, M.
Zenke, H. Beug, and M. L. Bimstiel. Transferrin-polycation-mediated
introduction of DNA into human leukemic cells: stimulation by agents that
affect the survival of transfected DNA or modulate transferrin receptor levels,
Proc. Natl. Acad. Sci. U. S. A, 87:4033-4037 (1990).
244. M. Zenke, P. Steinlein, E. Wagner, M. Gotten, H. Beug, and M. L. Bimstiel.
Receptor-mediated endocytosis of transferrin-polycation conjugates: an
efficient way to introduce DNA into hematopoietic cells, (1990).
245. E. Wagner, M. Zenke, M. Gotten, H. Beug, and M. L. Bimstiel. Transferrin-
polycation conjugates as carriers for DNA uptake into cells, Proc. Natl. Acad.
Sci. U. S. A, 87:3410-3414 (1990).
245
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
246. P. W. Cheng. Receptor ligand-facilitated gene transfer: enhancement of
liposome- mediated gene transfer and expression by transferrin, Hum. Gene
r/jer., 7:275-282 (1996).
247. S. Simoes, P. Pires, N. Duzgunes, and M. C. Pedrosa de Lima. Cationic
liposomes as gene transfer vectors: barriers to successful application in gene
therapy, Curr. Opin. Mol. Ther., 1:147-157 (1999).
248. M. C. de Lima, S. Simoes, P. Pires, R. Caspar, V. Slepushkin, and N.
Duzgunes. Gene delivery mediated by cationic liposomes: from biophysical
aspects to enhancement of transfection, Mol. Membr. B iol, 16:103-109
(1999).
249. K. Kono, Y. Torikoshi, M. Mitsutomi, T. Itoh, N. Emi, H. Yanagie, and T.
Takagishi. Novel gene delivery systems: complexes of flisigenic polymer-
modified liposomes and lipoplexes. Gene Ther. , 8:5-12 (2001).
250. K. Yanagihara, H. Cheng, and P. W. Cheng. Effects of epidermal growth
factor, transferrin, and insulin on lipofection efficiency in human Ixmg
carcinoma cells. Cancer Gene Ther., 7:59-65 (2000).
251. d. Xros, I and N. Duzgunes. Efficient gene transfer by transferrin lipoplexes
in the presence of serum, Biochim. Biophys. Acta, 1463:333-342 (2000).
252. S. Simoes, V. Slepushkin, R. Caspar, M. C. de Lima, and N. Duzgunes. Cene
delivery by negatively charged ternary complexes of DNA, cationic
liposomes and transferrin or fusigenic peptides. Gene Ther., 5:955-964
(1998).
253. H. linuma, K. Maruyama, K. Okinaga, K. Sasaki, T. Sekine, O. Ishida, N.
Ogiwara, K. Johkura, and Y. Yonemura. Intracellular targeting therapy of
cisplatin-encapsulated transferrin- polyethylene glycol liposome on
peritoneal dissemination of gastric cancer. Int. J. Cancer, 99:130-137 (2002).
246
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
254. A. G. Tonevitsky, I. A. Demina, 1.1. Agapov, G. V. Fattakhova, E. N.
Popova, M. M. Moysenovich, I. S. Komolov, A. S. Khalanskii, and M. P.
Kirpichnikov. Cytotoxic activities of conjugates of human transferrin and A
subunits of plant toxins both in vitro and in vivo, Dokl. Biochem., 374:210-
213 (2000).
255. C. Candiani, M. Tommasi, G. Fracasso, I. Lorenzetti, A. Adami, G. Benoni,
G. Tridente, and M. Colombatti. Pharmacokinetics of intrathecal transferrin-
ricin a chain immunotoxin, Life Sci, 69:335-346 (2001).
256. L. Chores, J. M. Ferreras, R. Munoz, J. Benitez, P. Jimenez, and T. Girbes.
Targeting cancer cells with transferrin conjugates containing the non- toxic
type 2 ribosome-inactivating proteins nigrin h or ehulin 1 , Cancer Lett.,
184:29-35 (2002).
257. S. B. Komfeld, J. E. Leonard, M. D. Mullen, and R. Taetle. Assessment of
ligand effects in intracellular trafficking of ricin A chain using anti-ricin
hybridomas. Cancer Res., 51:1689-1693 (1991).
258. N. Bejaoui, M. Page, and C. Noel. Cytotoxicity of transferrin-daunorubicin
conjugates on small cell carcinoma of the lung (SCCL) cell line NCI-H69,
Anticancer Res., 11:2211-2213 (1991).
259. D. W. Laske, R. J. Youle, and E. H. Oldfield. Tumor regression with regional
distribution of the targeted toxin TF- CRM 107 in patients with malignant
brain tumors, Nat. Med., 3:1362-1368 (1997).
260. N. Hagihara, S. Walbridge, A. W. Olson, E. H. Oldfield, and R. J. Youle.
Vascular protection by chloroquine during brain tumor therapy with Tf-
CRM107, Cancer Res., 60:230-234 (2000).
261. M. Singh, H. Atwal, and R. Micetich. Transferrin directed delivery of
adriamycin to human cells. Anticancer Res., 18:1423-1427 (1998).
262. M. Fritzer, T. Szekeres, V. Szuts, H. N. Jarayam, and H. Goldenberg.
Cytotoxic effects of a doxoruhicin-transferrin conjugate in multidrug-
resistant KB cells, Biochem. Pharmacol., 51:489-493 (1996).
247
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
263. L. Xu, K. F. Pirollo, and E. H. Chang. Transferrin-liposome-mediated p53
sensitization of squamous cell carcinoma of the head and neck to radiation in
vitro, Hum. Gene Ther., 8:467-475 (1997).
264. L. Xu, K. F. Pirollo, W. H. Tang, A. Rait, and E. H. Chang. Transferrin-
liposome-mediated systemic p53 gene therapy in combination with radiation
results in regression of human head and neck cancer xenografts. Hum. Gene
Ther., 10:2941-2952 (1999).
265. L. Xu, K. F. Pirollo, and E. H. Chang. Tumor-targeted p53-gene therapy
enhances the efficacy of conventional chemo/radiotherapy, J. Control
Release, 74:115-128(2001).
266. L. Xu, P. Frederik, K. F. Pirollo, W. H. Tang, A. Rait, L. M. Xiang, W.
Huang, I. Cruz, Y. Yin, and E. H. Chang. Self-assembly of a virus-mimicking
nanostructure system for efficient tumor-targeted gene delivery. Hum. Gene
Ther., 13:469-481 (2002).
267. M. Seki, J. Iwakawa, H. Cheng, and P. W. Cheng. p53 and
PTEN/MM AC 1 /TEP1 gene therapy of human prostate PC-3 carcinoma
xenograft, using transferrin-facilitated lipofection gene delivery strategy,
Hum. Gene Ther., 13:761-773 (2002).
268. E. Wagner, K. Zatloukal, M. Cotten, H. Kirlappos, K. Mechtler, D. T. Curiel,
and M. L. Bimstiel. Coupling of adenovims to transferrin-polylysine/DNA
complexes greatly enhances receptor-mediated gene delivery and expression
of transfected genes, Proc. Natl. Acad. Sci. U. S. A, 89:6099-6103 (1992).
269. G. Quick, J. van Zyl, A. Hawtrey, and M. Ariatti. Effect of nicotinic acid
conjugated to DNA-transfecting complexes targeted at the transferrin
receptor of HeLa cells. Drug Deliv., 7:231-236 (2000).
270. R. Kircheis, S. Schuller, S. Branner, M. Ogris, K. H. Heider, W. Zauner, and
E. Wagner. Polycation-based DNA complexes for tumor-targeted gene
delivery in vivo, J. Gene Med., 1:111-120 (1999).
248
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
271. R. Kircheis, L. Wightman, A. Schreiber, B. Robitza, V. Rossler, M. Kursa,
and E. Wagner. Poiyethylenimine/DNA complexes shielded by transferrin
target gene expression to tumors after systemic application, Gene Ther., 8:28-
40 (2001).
272. P. R. Dash, M. L. Read, K. D. Fisher, K. A. Howard, M. Wolfert, D.
Oupicky, V. Subr, J. Strohalm, K. Ulbrich, and L. W. Seymour. Decreased
binding to proteins and cells of polymeric gene delivery vectors surface
modified with a multivalent hydrophilic polymer and retargeting through
attachment of transferrin, J. Biol Chem., 275:3793-3802 (2000).
273. K. D. Fisher, K. Ulbrich, V. Subr, C. M. Ward, V. Mautner, D. Blakey, and
L. W. Seymour. A versatile system for receptor-mediated gene delivery
permits increased entry of DNA into target cells, enhanced delivery to the
nucleus and elevated rates of transgene expression. Gene Ther., 7:1337-1343
(2000).
274. H. A. Huehers and C. A. Finch. The physiology of transferrin and transferrin
receptors. Physiol Rev., 67:520-582 (1987).
275. J. H. Kordower, V. Charles, R. Bayer, R. T. Bartus, S. Putney, L. R. Walus,
and P. M. Friden. Intravenous administration of a transferrin receptor
antibody-nerve growth factor conjugate prevents the degeneration of
cholinergic striatal neurons in a model of Huntington disease, Proc. Natl.
Acad. Sci. U. S. A, 91:9077-9080 (1994).
276. P. M. Friden. Receptor-mediated transport of therapeutics across the blood-
brain barrier. Neurosurgery, 35:294-298 (1994).
277. U. Bickel, T. Yoshikawa, and W. M. Pardridge. Delivery of peptides and
proteins through the blood-brain barrier. Adv. Drug Deliv. Rev., 46:247-279
(2001).
278. T. Moos and E. H. Morgan. Restricted transport of anti-transferrin receptor
antibody (0X26) through the blood-brain barrier in the rat, J. Neurochem.,
79:119-129(2001).
249
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
279. W. M. Pardridge, J. L. Buciak, and P. M. Friden. Selective transport of an
anti-transferrin receptor antibody through the blood-brain barrier in vivo, J.
Pharmacol. Exp. Ther., 259:66-70 (1991).
280. U. Bickel, T. Yoshikawa, E. M. Landaw, K. F. Faull, and W. M. Pardridge.
Pharmacologic effects in vivo in brain by vector-mediated peptide drug
delivery, Proc. Natl. Acad. Sci. U. S. A, 90:2618-2622 (1993).
281. Y. Zhang and W. M. Pardridge. Neuroprotection in transient focal brain
ischemia after delayed intravenous administration of brain-derived
neurotrophic factor conjugated to a blood-brain barrier drug targeting system.
Stroke, 32:1378-1384 (2001).
282. Y. Zhang and W. M. Pardridge. Conjugation of brain-derived neurotrophic
factor to a blood-brain barrier drug targeting system enables neuroprotection
in regional brain ischemia following intravenous injection of the
neurotrophin. Brain Res., 889:49-56 (2001).
283. W. M. Pardridge, D. Wu, and T. Sakane. Combined use of carboxyl-directed
protein pegylation and vector- mediated blood-brain barrier drug delivery
system optimizes brain uptake of brain-derived neurotrophic factor following
intravenous administration, Pharm. Res., 15:576-582 (1998).
284. D. Wu, B. W. Song, H. V. Vinters, and W. M. Pardridge. Pharmacokinetics
and brain uptake of biotinylated basic fibroblast growth factor conjugated to a
blood-brain barrier drug delivery system, J. Drug Target, 10:239-245 (2002).
285. B. W. Song, H. V. Vinters, D. Wu, and W. M. Pardridge. Enhanced
neuroprotective effects of basic fibroblast growth factor in regional brain
ischemia after conjugation to a blood-brain barrier delivery vector, J.
Pharmacol. Exp. Ther., 301:605-610 (2002).
286. D. Wu and W. M. Pardridge. Neuroprotection with noninvasive neurotrophin
delivery to the brain, Proc. Natl. Acad. Sci. U. S. A, 96:254-259 (1999).
250
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
287. Y. Zhang and W. M. Pardridge. Mediated efflux of IgG molecules from brain
to blood across the blood- brain barrier, J NeuroimmunoL, 114:168-172
(2001).
288. Y. Zhang and W. M. Pardridge. Rapid transferrin efflux from brain to blood
across the blood-brain barrier, J. Neurochem., 76:1597-1600 (2001).
289. J. Huwyler, D. Wu, and W. M. Pardridge. Brain drug delivery of small
molecules using immunoliposomes, Proc. Natl. Acad. Sci. U. S. A, 93:14164-
14169 (1996).
290. J. Huwyler, A. Cerletti, G. Pricker, A. N. Eberle, and J. Drewe. By-passing of
P-glycoprotein using immunoliposomes, J. Drug Target, 10:73-79 (2002).
291. N. Shi, Y. Zhang, C. Zhu, R. J. Boado, and W. M. Pardridge. Brain-specific
expression of an exogenous gene after i.v. administration, Proc. Natl. Acad.
Sci. U. S. A, 98:12754-12759 (2001).
292. J. Wan, M. E. Taub, D. Shah, and W. C. Shen. Brefeldin A enhances
receptor-mediated transcytosis of transferrin in filter-grown Madin-Darby
canine kidney cells, J. Biol. Chem., 267:13446-13450 (1992).
293. W. C. Shen, D. Shah, M. Taub, and J. Wan. Enhancement of transferrin
receptor-mediated transcytosis for transepithelial protein delivery. In V. H. L.
Lee, M. Hashida, and Y. Mizushima (eds). Trends and future perspectives in
peptide and protein drug delivery., harwood academic publishers, 1995, pp.
135-151.
294. D. J. McCool, J. F. Forstner, and G. G. Forstner. Regulated and unregulated
pathways for MUC2 mucin secretion in human colonic LSI80
adenocarcinoma cells are distinct, Biochem. J , 312 ( Pt 1): 125-133 (1995).
295. P. P. Breitfeld, J. E. Casanova, N. E. Simister, S. A. Ross, W. C. McKinnon,
and K. E. Mostov. Sorting signals, Curr. Opin. Cell Biol., 1:617-623 (1989).
251
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
296. B. S. Stein and H. H. Sussman. Demonstration of two distinct transferrin
receptor recycling pathways and transferrin-independent receptor
internalization in K562 cells, J. Biol. Chem., 261:10319-10331 (1986).
297. R. D. Klausner, J. G. Donaldson, and J. Lippincott-Schuartz. Brefeldin A:
Insights into the control of membrane traffic and orgenelle structure., J. Cell
Biol, 116:1071-1080(1992).
298. J. Lippincott-Schuartz, L. Yuan, C. Tipper, M. Amherdt, L. Orci, and R. D.
Klausner. Brefeldin A's effects on endosomes, lysosomes and TON suggest a
gemal mechanism for regulating organelle structure and membrane traffic..
Cell, 67:601-616 (1991).
299. K. Oda, S. Hirose, N. Takami, A. Misumi, A. Takatsuki, and Y. Dcehara.
Brefedin A arrests the intracellular transport of a precursor of completement
C3 before its converson site in rat hepatocytes., FEBS Lett., 214:135-138
(1987).
300. A. Misumi, A. Miki, A. Takatsuki, G. Tamura, and Y. Dcehara. Novel
blockade by brefeldin A of intracellular transport of secretory proteins in rat
hepacytes., J. Biol Chem., 261:11398-11403 (1986).
301. J. Lippincott-Schuartz, J. G. Donaldson, A. J. G. Schweizer, E. G. Berger, H.
P. Hauri, L. C. Yuan, and R. D. Klausner. Microtube-dependent retrograde
transport of proteins into the ER in the presence of brefeldin A suggests an
ER recycling pathway.. Cell, 60:821-836 (1990).
302. R. Doms, G. Russ, and J. Yewdell. Brefeldin A redistributes resident and
itinerant Golgi proteins to endoplasmic reticulum., J. Cell Biol, 109:61-72
(1989).
303. S. Wood, J. Park, and W. Brown. Brefedin A causes a microtuble-mediated
fusion of the TGN and early endosomes.. Cell, 67:591-600 (1998).
304. K. Prydz, S. H. Hansen, K. Sandvig, and B. Van Deurs. Effects of brefeldin
A on endocytosis, transcytosis and transport to the Golgi complex in
polarized MDCk cells., J. Cell Biol, 119:259-272 (1992).
252
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
305. W. Hunziker, J. A. Whitney, and I. Mellman. Selective inhibition of
transcytosis by brefedin A in MDCK cells.. Cell, 67:617-627 (1991).
306. K. Matter, J. A. Whitney, E. M. Yamamoto, and I. Mellman. Common
signals control low density lipoprotein receptor sorting in endosomes and the
Golgi complex of MDCK cells. Cell, 74:1053-1064 (1993).
307. G. Apodaca, B. Aroeti, K. Tang, and K. Mostov. Brefeldin-A inhibits the
delivery of the polymeric immunoglobulin receptor to the basolateral surface
of MDCK cells., J. Biol. Chem., 268:20380-20385 (1993).
308. S. H. Low, B. L. Tang, S. H. Wong, and W. J. Hong. Selective inhibition of
protein targeting to the apical domain of MDCK cells by brefeldin A., J. Cell
Biol, 118 :51-62 (1992).
309. M. Wagner, A. K. Rajasekaran, D. K. Hanzel, S. Mayor, and E. Rodriguez-
Boulan. Brefeldin A causes structural and functional alterations of trans-
Golgi network of MDCK cells., J. Cell Sci., 107:933-943 (1994).
310. Widera A., Kim K.J., Crandall E.D., and Shen W.-C. Transcytosis of GCSF-
Transferrin Across Rat Alveolar Epithelial Cell Monolayers, Pharm. Res., (in
press): (2003).
311. J. Wang, D. Shen, and W. C. Shen. Oral delivery of insulin-transferrin
conjugate in streptozotocin-treated CF/1 mice, Pharm. Res. (Supplement),
14:469 (1997).
312. C. Q. Xia and W. C. Shen. Tyrphostin-8 enhances transferrin receptor-
mediated transcytosis in Caco-2- cells and inreases hypoglycemic effect of
orally administered insulin-transferrin conjugate in diabetic rats, Pharm. Res.,
18:191-195(2001).
313. G. Li, C. D'Souza-Schorey, M. A. Barbieri, R. L. Roberts, A. Klippel, L. T.
Williams, and P. D. Stahl. Evidence for phosphatidylinositol 3-kinase as a
regulator of endocytosis via activation of Rab5, Proc. Natl. Acad. Sci. U. S.
A, 92:10207-10211 (1995).
253
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
314. D. J. Spiro, W. Boll, T. Kirchhausen, and M. Wessling-Resnick. Wortmannin
alters the transferrin receptor endocytic pathway in vivo and in vitro. Mol.
Biol. Cell, 7:355-367 (1996).
315. P. R. Shepherd, M. A. Soos, and K. Siddle. Inhibitors of phosphoinositide 3-
kinase block exocytosis but not endocytosis of transferrin receptors in 3T3-
L1 adipocytes, Biochem. Biophys. Res. Commun., 211:535-539 (1995).
316. K. W. Ko, R. K. Avramoglu, R. S. McLeod, J. Vukmirica, and Z. Yao. The
insulin-stimulated cell surface presentation of low density lipoprotein
receptor-related protein in 3T3-L1 adipocytes is sensitive to
phosphatidylinositide 3-kinase inhibition, Biochemistry, 40:752-759 (2001).
317. J. M. Gutteridge, S. Mumby, G. J. Quinlan, K. F. Chung, and T. W. Evans.
Pro-oxidant iron is present in human pulmonary epithelial lining fluid:
implications for oxidative stress in the Ixmg, Biochem. Biophys. Res.
Commun., 220:1024-1027 (1996).
318. K. R. Smith and A. E. Aust. Mobilization of iron from urban particulates
leads to generation of reactive oxygen species in vitro and induction of
ferritin synthesis in human limg epithelial cells, Chem. Res. Toxicol, 10:828-
834(1997).
319. O. Olakanmi, S. E. McGowan, M. B. Hayek, and B. E. Britigan. Iron
sequestration by macrophages decreases the potential for extracellular
hydroxyl radical formation, J. Clin. Invest, 91:889-899 (1993).
320. C. E. Alford, T. E. King, Jr., and P. A. Campbell. Role of transferrin,
transferrin receptors, and iron in macrophage listericidal activity, J. Exp.
Med., 174:459-466 (1991).
321. J. J. Bullen and J. A. Armstrong. The role of lactoferrin in the bactericidal
function of polymorphonuclear leucocytes. Immunology, 36:781-791 (1979).
254
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
322. B. E. Britigan, M. B. Hayek, B. N. Doebbeling, and R. B. Pick, Jr.
Transferrin and lactoferrin undergo proteolytic cleavage in the Pseudomonas
aeruginosa-infected lungs of patients with cystic fibrosis. Infect. Immun.,
61:5049-5055 (1993).
323. A. B. Thompson, T. Bohling, A. Heires, J. Linder, and S. I. Rennard. Lower
respiratory tract iron burden is increased in association with cigarette
smoking, J. Lab Clin. Med., 117:493-499 (1991).
324. H. A. Huebers and C. A. Finch. The physiology of transferrin and transferrin
receptors. Physiol Rev., 67:520-582 (1987).
325. E. R. Pacht and W. B. Davis. Role of transferrin and ceruloplasmin in
antioxidant activity of lung epithelial lining fluid, J. Appl. Physiol, 64:2092-
2099 (1988).
326. D. Y. Bell, J. A. Haseman, A. Spock, G. McLennan, and G. E. Hook. Plasma
proteins of the bronchoalveolar surface of the lungs of smokers and
nonsmokers. Am. Rev. Respir. Dis., 124:72-79 (1981).
327. S. W. Stites, M. E. Nelson, and L. J. Wesselius. Transferrin concentrations in
serum and lower respiratory tract fluid of mechanically ventilated patients
with COPD or ARDS, Chest, 107:1681-1685 (1995).
328. J. F. Holter, J. E. Weiland, E. R. Pacht, J. E. Gadek, and W. B. Davis. Protein
permeability in the adult respiratory distress syndrome. Loss of size
selectivity of the alveolar epithelium, J. Clin. Invest, 78:1513-1522 (1986).
329. J. M. Cheek, M. J. Evans, and E. D. Crandall. Type I cell-like morphology in
tight alveolar epithelial monolayers, Exp. Cell Res., 184:375-387 (1989).
330. L. G. Dobbs, M. C. Williams, and R. Gonzalez. Monoclonal antibodies
specific to apical surfaces of rat alveolar type I cells bind to surfaces of
cultured, but not freshly isolated, type II cells, Biochim. Biophys. Acta,
970:146-156(1988).
255
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
331. J. M. Shannon, S. D. Jennings, and L. D. Nielsen. Modulation of alveolar
type n cell differentiated function in vitro. Am. J. Physiol, 262:L427-L436
(1992).
332. S. I. Danto, J. M. Shannon, Z. Borok, S. M. Zabski, and E. D. Crandall.
Reversible transdifferentiation of alveolar epithelial cells. Am. J. Respir. Cell
Mol. Biol, 12:497-502 (1995).
333. Z. Borok, R. L. Lubman, S. I. Danto, X. L. Zhang, S. M. Zabski, L. S. King,
D. M. Lee, P. Agre, and E. D. Crandall. Keratinocyte growth factor
modulates alveolar epithelial cell phenotype in vitro: expression of aquaporin
5, Am. J. Respir. Cell M ol Biol, 18:554-561 (1998).
334. Z. Borok, S. I. Danto, R. L. Lubman, Y. Cao, M. C. Williams, and E. D.
Crandall. Modulation of tl alpha expression with alveolar epithelial cell
phenotype in vitro, Am. J. Physiol, 275:L155-L164 (1998).
335. B. E. Isakson, R. L. Lubman, G. J. Seedorf, and S. Boitano. Modulation of
pulmonary alveolar type II cell phenotype and communication by
extracellular matrix and KGF, Am. J. Physiol Cell Physiol, 281:C1291-C1299
(2001).
336. K. Sugahara, J. S. Rubin, R. J. Mason, E. L. Aronsen, and J. M. Shannon.
Keratinocyte growth factor increases mRNAs for SP-A and SP-B in adult rat
alveolar type II cells in culture. Am. J. Physiol, 269:L344-L350 (1995).
337. Azari, K. L. and Feeney, R. E. Resistance of metal complexes of conalbumin
and transferrin to proteolysis and thermal denaturation. J.Biol.Chem. 232,
293-302. 1958.
Ref Type: Generic
338. D. Banerjee, P. R. Flanagan, J. Cluett, and L. S. Valberg. Transferrin
receptors in the human gastrointestinal tract. Relationship to body iron stores.
Gastroenterology, 91:861-869 (1986).
339. R. R. Crichton. Proteins of iron storage and transport, Adv. Protein Chem.,
40:281-363 (1990).
256
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
340. H. O'Brodovich, C. Canessa, J. Ueda, B. Rafii, B. C. Rossier, and J. Edelson.
Expression of the epithelial Na+ channel in the developing rat lung, Am. J.
Physiol, 265:C491-C496 (1993).
341. K. Matsushita, P. B. McCray, Jr., R. D. Sigmund, M. J. Welsh, and J. B.
Stokes. Localization of epithelial sodium channel subunit mRNAs in adult rat
lung by in situ hybridization. Am. J. Physiol, 271:L332-L339 (1996).
342. Z. Borok, J. M. Liebler, R. L. Lubman, M. J. Foster, B. Zhou, X. Li, S. M.
Zabski, K. J. Kim, and E. D. Crandall. Na transport proteins are expressed by
rat alveolar epithelial type I cells. Am. J. Physiol Lung Cell Mol. Physiol,
282:L599-L608 (2002).
343. Widera A., Kim K.J., Crandall E.D., and Shen W.-C. Transcytosis of GCSF-
Transferrin Across Rat Alveolar Epithelial Cell Monolayers, Pharm. Res., (in
press): (2003).
344. Z. Borok, S. I. Danto, S. M. Zabski, and E. D. Crandall. Defined medium for
primary culture de novo of adult rat alveolar epithelial cells. In Vitro Cell
Dev. Biol. Anim, 30A:99-104 (1994).
345. S. Sonoda and M. Schlamowitz. Studies of 1251 trace labeling of
immunoglobulin G by chloramine-T, Immunochemistry., 7:885-898 (1970).
346. J. E. Lamb, F. Ray, J. H. Ward, J. P. Kushner, and J. Kaplan. Intemalization
and subcellular localization of transferrin and transferrin receptors in HeLa
cells, J. Biol. Chem., 258:8751-8758 (1983).
347. L. M. Timchak, F. Kruse, M. H. Mamell, and R. K. Draper. A
thermosensitive lesion in a Chinese hamster cell mutant causing differential
effects on the acidification of endosomes and lysosomes, J. Biol. Chem.,
261:14154-14159(1986).
348. L. M. Souza, T. C. Boone, J. Gabrilove, P. H. Lai, K. M. Zsebo, D. C.
Murdock, V. R. Chazin, J. Bruszewski, H. Lu, K. K. Chen, and .
Recombinant human granulocyte colony-stimulating factor: effects on normal
and leukemic myeloid cells. Science, 232:61-65 (1986).
257
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
349. N. Shirafuji, S. Asano, S. Matsuda, K. Watari, F. Takaku, and S. Nagata. A
new bioassay for human granulocyte colony-stimulating factor (hG-CSF)
using murine myeloblastic NFS-60 cells as targets and estimation of its levels
in sera from normal healthy persons and patients with infectious and
hematological disorders, Exp. Hematol., 17:116-119 (1989).
350. T. Mosmann. Rapid colorimetric assay for cellular growth and survival:
application to proliferation and cytotoxicity assays, J. Immunol. Methods,
65:55-63 (1983).
351. F. Heckner. Practical Microscopic Hematology, Urban & Schwarzenberg,
Baltimore, 1982.
352. W. M. Pardridge, J. L. Buciak, and P. M. Friden. Selective transport of an
anti-transferrin receptor antibody through the blood-brain barrier in vivo, J.
Pharmacol. Exp. Ther., 259:66-70 (1991).
353. J. Wan, M. Taub, D. Shah, and W. C. Shen. Brefeldin A enhances receptor-
mediated transcytosis of Tf in filter grown Madin-darby canine kidney cells.,
J. Biol. Chem., 267:13446-13450 (1992).
354. D. Shah and W. C. Shen. The establishment of polarity and enhancd
transcytosis of transferrin receptors in enterocyte-like Caco-2 cells., J. Drug
Targeting, 2:93-99 (1994).
355. P. E. Thorpe, P. Wallace, P. Knowles, M. Relf, A. Brown, G. Waston, D.
Blakey, and D. Newell. Improved antitumor effects of immunotoxins
prepared with deglycosylated ricin A-chain and hindered disulfide linkages.
Cancer Res., 48:6396-6403 (1988).
356. D. Banerjee, P. R. Flanagan, J. Cluett, and L. S. Valberg. Transferrin
receptors in the human gastrointestinal tract., Gastroenterol, 91:861-869
(1986).
357. A. Dautry-Varsat. Receptor-mediated endocytosis: the intracellular journey
of transferrin and its receptor., Biochimie, 68:375-381 (1986).
258
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
358. A. H. Le Gall, C. Yeaman, A. Muesch, and E. Rodriguez-Boulan. Epithelial
cell polarity: new perspectives., Seminars in Nephrology, 15:272-284 (1995).
359. K. Simons and A. Wandinger-Ness. Polarized sorting in epithelia.. Cell,
62:207-210 (1990).
360. A. Le Bivic, A. Quaroni, B. Nichols, and E. Rodriguez-Boulan. Biogenetic
pathways of plasma membrane proteins in Caco-2, a human intestinal
epithelial cell line.,/. Cell Biol, 111:1351-1361 (1990).
361. K. Matter, M. Brauchbar, K. Bucher, and H. P. Hauri. Sorting of endogenous
plasma membrane proteins occurs from two sites in cutured human intestinal
epithelial cells (Caco-2)., Cell, 60:429-437 (1990).
362. H. Damke, J. Klumperman, K. von Figura, and T. Braulke. Brefeldin A
affects the cellular distribution of endocytic receptors differentially, Biochem.
Biophys. Res. Commun., 185:719-727 (1992).
363. R. D. Klausner, G. Ashwell, J. van Renswoude, J. B. Harford, and K. R.
Bridges. Binding of apotransferrin to K562 cells: explanation of the
transferrin cycle, Proc. Natl. Acad. Sci. U. S. A , 80:2263-2266 (1983).
364. T. Fujiwara, K. Oda, S. Yokota, A. Takatsuki, and Y. Ikehara. Brefeldin A
causes disassembly of the Golgi complex and accumulation of secretory
proteins in the endoplasmic reticulum, J. Biol. Chem., 263:18545-18552
(1988).
365. J. B. Ulmer and G. E. Palade. Targeting and processing of glycophorins in
murine erythroleukemia cells: use of brefeldin A as a perturbant of
intracellular traffic, Proc. Natl. Acad. Sci. U . S. A, 86:6992-6996 (1989).
366. J. B. Ulmer and G. E. Palade. Effects of brefeldin A on the processing of
viral envelope glycoproteins in murine erythroleukemia cells, J. Biol Chem.,
266:9173-9179 (1991).
259
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
367. D. Deshpande, D. Toledo-Velasquez, L. Y. Wang, C. J. Malanga, J. K. Ma,
and Y. Rojanasakul. Receptor-mediated peptide delivery in pulmonary
epithelial monolayers, Pharm. Res., 11:1121-1126 (1994).
368. I. J. Hidalgo, T. J. Raub, and R. T. Borchardt. Characterization of the human
colon carcinoma cell line (Caco-2) as a model system for intestinal epithelial
permeability. Gastroenterology, 96:736-749 (1989).
369. K. L. Audus, R. L. Bartel, I. J. Hidalgo, and R. T. Borchart. The use of
cultured epithelial and endothelial cells for drug transport and metabolism
studies., Pharm. Res., 7:435-451 (1990).
370. A. Quaroni and J. Hochman. Development of intestinal cell culture models
for drug transport and metabolism studies.. Adv. Drug Delivery Rev., 22:3-52
(1996).
371. L. L. Gan and D. R. Thakker. Application of the Caco-2 model in the design
and development of orally active drugs: elucidation of biochemical and
physiological barriers posed by the intestinal epithium.. Adv. Drug Delivery
Rev., 23:77-98 (1997).
372. Kuwabara T., Kato Y., Kobayashi S., Suzuki H., and Sugiyama Y. Nonlinear
pharmacokinetics of a recombinant human granulocyte colony-stimulating
factor derivative (nartograstim): species differences among rats, monkeys and
humans., J. Pharmacol. Exp. Ther., 271:1535-1543 (1994).
373. W. Halpem, T. A. Riccobene, H. Agostini, K. Baker, D. Stolow, M. L. Gu, J.
Hirsch, A. Mahoney, J. Carrell, E. Boyd, and K. J. Grzegorzewski.
Albugranin, a recombinant human granulocyte colony stimulating factor (G-
CSF) genetically fused to recombinant human albumin induces prolonged
myelopoietic effects in mice and monkeys, Pharm. Res., 19:1720-1729
(2002).
374. B. I. Lord, L. B. Woolford, and G. Molineux. Kinetics of neutrophil
production in normal and neutropenic animals during the response to
filgrastim (r-metHu G-CSF) or filgrastim SD/01 (PEG-r-metHu G-CSF),
Clin. Cancer Res., 7:2085-2090 (2001).
260
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
375. L. Li and J. Kaplan. Alteration in the organ distribution of iron by truncated
transferrin: implications for iron chelation therapy, J. Laboratory & Clinical
Medicine, 130:271-277 (1997).
376. Tanaka H. and Kaneko T. Pharmacokinetics of recombinant human
granulocyte colony-stimulating factor in mice., Blood, 79:536-539 (1992).
377. M. A. Winkler, J. O. Price, P. D. Foglesong, and W. H. West. Biodistribution
and plasma survival in mice of anti-melanoma monoclonal antibody cross-
linked to 0KT3., Cancer Immunology, Immunotherapy., 31:278-284 (1990).
378. C. N. Roy and C. A. Enns. Iron homeostasis: new tales from the crypt. Blood,
96:4020-4027 (2000).
261
Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.
Linked assets
University of Southern California Dissertations and Theses
Conceptually similar
PDF
Pharmacological regulation of chloride, fluid, and solute transport in the pigmented rabbit conjunctiva
PDF
Modulation of macrophage glutathione synthesis by oxidized LDL and the effects of glutathione content on cell-mediated LDL oxidation
PDF
Nonreceptor tyrosine kinase Etk /Bmx: Function and regulation in epithelial cells
PDF
The mechanism for the regulation of RNA polymerase I-dependent transcription
PDF
Structure-function studies on the mammalian intestinal dipeptide transporter hPEPT1
PDF
Organic cation transport in the cultured alveolar epithelial cells: Functional and molecular studies
PDF
Role of PKC-delta in deferoxamine-induced cell death in salivary epithelial Pa-4 cells
PDF
Glutathione metabolism and transport in cystic fibrosis airway epithelia
PDF
Basolateral transport of nucleosides in rabbit tracheal epithelial cells: An equilibrative nucleoside transporter outcome
PDF
Uptake and transport of desmopressin and its lipidized derivatives in Caco-2 cells
PDF
Discovery of a novel role of vasopressin in astrocytes: Vasopressin-induced cytoplasmic and nuclear calcium and kinases signaling cascade and modulation of astrocytes immune function
PDF
A dynamic apical actin cytoskeleton facilitates exocytosis of tear proteins in rabbit lacrimal acinar epithelial cells
PDF
Studies on the mechanism and regulation of the transport of 5-fluorouracil (5FU) into tumor cells
PDF
An updated review of intracellular mRNA localization
PDF
Transferrin and its oligomer: Intracellular processing and applications as drug carriers
PDF
TfR-mediated oral delivery of protein drugs: Oral delivery of recombinant G -CSF -Tf fusion protein and its spacer optimization
PDF
Localization, sorting, and regulation of PepT1
PDF
The structure-function relationship of the human intestinal di-peptide transporter
PDF
Homeostatic regulation of intracellular signaling networks by Etk /Bmx
PDF
Preferential translation of Drosophila heat shock protein 70 and heat shock protein 90 mRNAs during heat shock
Asset Metadata
Creator
Widera, Adam Scott (author)
Core Title
Trans-epithelial transport of G -CSF -transferrin conjugate
Degree
Doctor of Philosophy
Degree Program
Pharmaceutical Sciences
Publisher
University of Southern California
(original),
University of Southern California. Libraries
(digital)
Tag
biology, cell,biology, molecular,Health Sciences, Pharmacology,OAI-PMH Harvest
Language
English
Contributor
Digitized by ProQuest
(provenance)
Permanent Link (DOI)
https://doi.org/10.25549/usctheses-c16-531455
Unique identifier
UC11336058
Identifier
3140572.pdf (filename),usctheses-c16-531455 (legacy record id)
Legacy Identifier
3140572.pdf
Dmrecord
531455
Document Type
Dissertation
Rights
Widera, Adam Scott
Type
texts
Source
University of Southern California
(contributing entity),
University of Southern California Dissertations and Theses
(collection)
Access Conditions
The author retains rights to his/her dissertation, thesis or other graduate work according to U.S. copyright law. Electronic access is being provided by the USC Libraries in agreement with the au...
Repository Name
University of Southern California Digital Library
Repository Location
USC Digital Library, University of Southern California, University Park Campus, Los Angeles, California 90089, USA
Tags
biology, cell
biology, molecular
Health Sciences, Pharmacology