Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters











Publication year range
1.
Traffic ; 2(3): 167-74, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11260522

ABSTRACT

People suffering from hereditary hemochromatosis (HH) can not regulate the uptake of iron properly and gradually accumulate iron in their body over their lifetime. The protein involved in HH, HFE, has been recently identified as a class I major histocompatibility complex (MHC) homolog. The wild-type HFE associates and co-traffics with the transferrin receptor (TfR). The mutation responsible for 83% of HH (C260Y) results in the failure of HFE to form a critical disulfide bond, bind beta2 microglobulin, bind TfR, and traffic to the cell surface. In non-polarized cells, the partnership of HFE and TfR results in decreased iron uptake into cells. The mechanism whereby a class I MHC homolog modifies the function of a membrane receptor and how this dynamic complex of molecules regulates iron transport across intestinal epithelial cells is the subject of this review.


Subject(s)
HLA Antigens/metabolism , Hemochromatosis/metabolism , Histocompatibility Antigens Class I/metabolism , Iron/metabolism , Membrane Proteins , Receptors, Transferrin/metabolism , Animals , HLA Antigens/chemistry , HLA Antigens/genetics , Hemochromatosis/genetics , Hemochromatosis Protein , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/physiology , Homeostasis , Humans , Models, Biological , Protein Structure, Secondary , Receptors, Transferrin/chemistry , beta 2-Microglobulin/chemistry , beta 2-Microglobulin/metabolism
2.
Blood ; 96(13): 4020-7, 2000 Dec 15.
Article in English | MEDLINE | ID: mdl-11110669

ABSTRACT

The enterocyte is a highly specialized cell of the duodenal epithelium that coordinates iron uptake and transport into the body. Until recently, the molecular mechanisms underlying iron absorption and iron homeostasis have remained a mystery. This review focuses on the proteins and regulatory mechanisms known to be present in the enterocyte precursor cell and in the mature enterocyte. The recent cloning of a basolateral iron transporter and investigations into its regulation provide new insights into possible mechanisms for iron transport and homeostasis. The roles of proteins such as iron regulatory proteins, the hereditary hemochromatosis protein (HFE)-transferrin receptor complex, and hephaestin in regulating this transporter and in regulating iron transport across the intestinal epithelium are discussed. A speculative, but testable, model for the maintenance of iron homeostasis, which incorporates the changes in the iron-related proteins associated with the life cycle of the enterocyte as it journeys from the crypt to the tip of the villous is proposed.


Subject(s)
Cation Transport Proteins , Intestinal Mucosa/metabolism , Iron/metabolism , Anemia, Hypochromic/genetics , Anemia, Hypochromic/metabolism , Biological Transport , Carrier Proteins/metabolism , Cell Differentiation , Cell Polarity , Duodenum/metabolism , Epithelial Cells/metabolism , Ferritins/metabolism , HLA Antigens/genetics , HLA Antigens/metabolism , Hemochromatosis/genetics , Hemochromatosis/metabolism , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Homeostasis , Humans , Intestinal Absorption , Intestinal Mucosa/ultrastructure , Iron/pharmacokinetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microvilli/metabolism , Models, Biological , Protein Transport , Receptors, Transferrin/metabolism , Stem Cells/metabolism , Transferrin/metabolism
3.
FEBS Lett ; 484(3): 271-4, 2000 Nov 10.
Article in English | MEDLINE | ID: mdl-11078891

ABSTRACT

Expression of wild type HFE reduces the ferritin levels of cells in culture. In this report we demonstrate that the predominant hereditary hemochromatosis mutation, C282Y(2) HFE, does not reduce ferritin expression. However, the second mutation, H63D HFE, reduces ferritin expression to a level indistinguishable from cells expressing wild type HFE. Further, two HFE cytoplasmic domain mutations engineered to disrupt potential signal transduction, S335M and Y342C, were functionally indistinguishable from wild type HFE in this assay, as was soluble HFE. These results implicate a role for the interaction of HFE with the transferrin receptor in lowering cellular ferritin levels.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Iron/metabolism , Membrane Proteins , Receptors, Transferrin/metabolism , Cell Line , HLA Antigens/chemistry , HeLa Cells , Hemochromatosis/genetics , Hemochromatosis Protein , Histocompatibility Antigens Class I/chemistry , Homeostasis , Humans , Receptors, Transferrin/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
4.
J Biol Chem ; 275(49): 38135-8, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11027676

ABSTRACT

The transferrin receptor (TfR) interacts with two proteins important for iron metabolism, transferrin (Tf) and HFE, the protein mutated in hereditary hemochromatosis. A second receptor for Tf, TfR2, was recently identified and found to be functional for iron uptake in transfected cells (Kawabata, H., Germain, R. S., Vuong, P. T., Nakamaki, T., Said, J. W., and Koeffler, H. P. (2000) J. Biol. Chem. 275, 16618-16625). TfR2 has a pattern of expression and regulation that is distinct from TfR, and mutations in TfR2 have been recognized as the cause of a non-HFE linked form of hemochromatosis (Camaschella, C., Roetto, A., Cali, A., De Gobbi, M., Garozzo, G., Carella, M., Majorano, N., Totaro, A., and Gasparini, P. (2000) Nat. Genet. 25, 14-15). To investigate the relationship between TfR, TfR2, Tf, and HFE, we performed a series of binding experiments using soluble forms of these proteins. We find no detectable binding between TfR2 and HFE by co-immunoprecipitation or using a surface plasmon resonance-based assay. The affinity of TfR2 for iron-loaded Tf was determined to be 27 nm, 25-fold lower than the affinity of TfR for Tf. These results imply that HFE regulates Tf-mediated iron uptake only from the classical TfR and that TfR2 does not compete for HFE binding in cells expressing both forms of TfR.


Subject(s)
HLA Antigens/chemistry , HLA Antigens/metabolism , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Membrane Proteins , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , Transferrin/chemistry , Transferrin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Dimerization , Hemochromatosis/genetics , Hemochromatosis Protein , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
5.
Nat Cell Biol ; 2(12): 953-7, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11146662

ABSTRACT

HFE, the protein that is mutated in hereditary haemochromatosis, binds to the transferrin receptor (TfR). Here we show that wild-type HFE and TfR localize in endosomes and at the basolateral membrane of a polarized duodenal epithelial cell line, whereas the primary haemochromatosis HFE mutant, and another mutant with impaired TfR-binding ability accumulate in the ER/Golgi and at the basolateral membrane, respectively. Levels of the iron-storage protein ferritin are greatly reduced and those of TfR are slightly increased in cells expressing wild-type HFE, but not in cells expressing either mutant. Addition of an endosomal-targeting sequence derived from the human low-density lipoprotein receptor (LDLR) to the TfR-binding-impaired mutant restores its endosomal localization but not ferritin reduction or TfR elevation. Thus, binding to TfR is required for transport of HFE to endosomes and regulation of intracellular iron homeostasis, but not for basolateral surface expression of HFE.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Iron/metabolism , Membrane Proteins , Receptors, Transferrin/metabolism , Biological Transport, Active , Cell Line , Cell Polarity , Endocytosis , Endosomes/metabolism , Epithelial Cells/metabolism , Green Fluorescent Proteins , HLA Antigens/genetics , Hemochromatosis/genetics , Hemochromatosis/immunology , Hemochromatosis/metabolism , Hemochromatosis Protein , Histocompatibility Antigens Class I/genetics , Homeostasis , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Models, Biological , Mutation , Protein Binding , Receptors, LDL/genetics , Receptors, LDL/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection
6.
Cell Microbiol ; 2(3): 207-18, 2000 Jun.
Article in English | MEDLINE | ID: mdl-11207577

ABSTRACT

Iron is an essential element for nearly all organisms. In mammals, iron is transported to body tissues by the serum glycoprotein transferrin. Transferrin-iron is internalized by binding to specific receptors followed by endocytosis. In vitro, Neisseria meningitidis and Neisseria gonorrhoeae can use iron from a variety of iron-containing compounds, including human transferrin. In vivo, transferrin is an important source of iron for N. gonorrhoeae: a mutant that is unable to bind and use transferrin-iron is unable to colonize the urethra of men or initiate disease at this site. As pathogenic Neisseria and its human host derive much of their iron from transferrin, we reasoned that a competition may exist between microbe and host epithelial cells for transferrin-iron at certain stages of infection. We therefore tested the hypothesis that N. meningitidis and N. gonorrhoeae may actively interfere with host transferrin-iron metabolism. We report that Neisseria-infected human epithelial cells have reduced levels of transferrin receptor messenger RNA and cycling transferrin receptors. The ability of infected cells to internalize transferrin receptor is also reduced. Finally, the relative distribution of surface and cycling transferrin receptors is altered in an infected cell. We conclude that Neisseria infection alters epithelial cell transferrin-iron homeostasis at multiple levels.


Subject(s)
Epithelial Cells/microbiology , Iron/metabolism , Neisseria gonorrhoeae/pathogenicity , Neisseria meningitidis/pathogenicity , Transferrin/metabolism , Gonorrhea/microbiology , Gonorrhea/physiopathology , Homeostasis , Humans , Meningococcal Infections/microbiology , Meningococcal Infections/physiopathology , RNA, Messenger/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Tumor Cells, Cultured
7.
J Immunol ; 163(1): 11-4, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10384092

ABSTRACT

Several studies on disposal of nonsecreted Ig L chains have identified the endoplasmic reticulum as the site of degradation. Here, we examine degradation of a nonsecreted Ig L chain, T15L, and an experimentally endoplasmic reticulum-retained secretion-competent L chain, D16L, in the absence of H chains. We demonstrate that 1) degradation is specifically impaired by the proteasome-specific inhibitors carboxybenzyl-leucyl-leucyl-leucine vinyl sulfone (Z-L3VS) and lactacystin, 2) L chain degradation occurs early in the biosynthetic pathway, and 3) degradation does not require vesicular transport. Our findings indicate that previous assertions of L chain disposal within the endoplasmic reticulum must be modified. To our knowledge, we provide the first direct evidence supporting a new paradigm for removal of nonsecreted Ig L chains via dislocation to cytosolic proteasomes.


Subject(s)
Cysteine Endopeptidases/physiology , Immunoglobulin Light Chains/metabolism , Multienzyme Complexes/physiology , Protein Processing, Post-Translational/immunology , Animals , Biological Transport/drug effects , Biological Transport/immunology , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/enzymology , Cytoplasm/immunology , Cytoplasm/metabolism , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/enzymology , Golgi Apparatus/immunology , Immunoglobulin Idiotypes/metabolism , Lysosomes/drug effects , Lysosomes/immunology , Lysosomes/metabolism , Mice , Multienzyme Complexes/immunology , Multienzyme Complexes/metabolism , Multiple Myeloma , Myeloma Proteins/metabolism , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational/drug effects , Tumor Cells, Cultured
8.
Mol Microbiol ; 32(6): 1316-32, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10383771

ABSTRACT

The pathogenic Neisseriae Neisseria meningitidis and Neisseria gonorrhoeae, initiate colonization by attaching to host cells using type IV pili. Subsequent adhesive interactions are mediated through the binding of other bacterial adhesins, in particular the Opa family of outer membrane proteins. Here, we have shown that pilus-mediated adhesion to host cells by either meningococci or gonococci triggers the rapid, localized formation of dramatic cortical plaques in host epithelial cells. Cortical plaques are enriched in both components of the cortical cytoskeleton and a subset of integral membrane proteins. These include: CD44v3, a heparan sulphate proteoglycan that may serve as an Opa receptor; EGFR, a receptor tyrosine kinase; CD44 and ICAM-1, adhesion molecules known to mediate inflammatory responses; f-actin; and ezrin, a component that tethers membrane components to the actin cytoskeleton. Genetic analyses reveal that cortical plaque formation is highly adhesin specific. Both pilE and pilC null mutants fail to induce cortical plaques, indicating that neisserial type IV pili are required for cortical plaque induction. Mutations in pilT, a gene required for pilus-mediated twitching motility, confer a partial defect in cortical plaque formation. In contrast to type IV pili, many other neisserial surface structures are not involved in cortical plaque induction, including Opa, Opc, glycolipid GgO4-binding adhesins, polysialic acid capsule or a particular lipooligosaccharide variant. Furthermore, it is shown that type IV pili allow gonococci to overcome the inhibitory effect of heparin, a soluble receptor analogue, on gonococcal invasion of Chang and A431 epithelial cells. These and other observations strongly suggest that type IV pili play an active role in initiating neisserial infection of the mucosal surface in vivo. The functions of type IV pili and other neisserial adhesins are discussed in the specific context of the mucosal microenvironment, and a multistep model for neisserial colonization of mucosal epithelia is proposed.


Subject(s)
Neisseria gonorrhoeae/pathogenicity , Neisseria meningitidis/pathogenicity , Pili, Sex/physiology , Animals , Cell Membrane , Cytochalasin D/pharmacology , Cytoskeleton/physiology , Epithelial Cells/cytology , Heparin/pharmacology , Humans , Membrane Proteins/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Rabbits , Tumor Cells, Cultured
9.
J Biol Chem ; 274(13): 9022-8, 1999 Mar 26.
Article in English | MEDLINE | ID: mdl-10085150

ABSTRACT

HFE is the protein product of the gene mutated in the autosomal recessive disease hereditary hemochromatosis (Feder, J. N., Gnirke, A., Thomas, W., Tsuchihashi, Z., Ruddy, D. A., Basava, A., Dormishian, F., Domingo, R. J., Ellis, M. C., Fullan, A., Hinton, L. M., Jones, N. L., Kimmel, B. E., Kronmal, G. S., Lauer, P., Lee, V. K., Loeb, D. B., Mapa, F. A., McClelland, E., Meyer, N. C., Mintier, G. A., Moeller, N., Moore, T., Morikang, E., Prasss, C. E., Quintana, L., Starnes, S. M., Schatzman, R. C., Brunke, K. J., Drayna, D. T., Risch, N. J., Bacon, B. R., and Wolff, R. R. (1996) Nat. Genet. 13, 399-408). At the cell surface, HFE complexes with transferrin receptor (TfR), increasing the dissociation constant of transferrin (Tf) for its receptor 10-fold (Gross, C. N., Irrinki, A., Feder, J. N., and Enns, C. A. (1998) J. Biol. Chem. 273, 22068-22074; Feder, J. N., Penny, D. M., Irrinki, A., Lee, V. K., Lebron, J. A., Watson, N. , Tsuchihashi, Z., Sigal, E., Bjorkman, P. J., and Schatzman, R. C. (1998) Proc. Natl. Acad. Sci. U S A 95, 1472-1477). HFE does not remain at the cell surface, but traffics with TfR to Tf-positive internal compartments (Gross et al., 1998). Using a HeLa cell line in which the expression of HFE is controlled by tetracycline, we show that the expression of HFE reduces 55Fe uptake from Tf by 33% but does not affect the endocytic or exocytic rates of TfR cycling. Therefore, HFE appears to reduce cellular acquisition of iron from Tf within endocytic compartments. HFE specifically reduces iron uptake from Tf, as non-Tf-mediated iron uptake from Fe-nitrilotriacetic acid is not altered. These results explain the decreased ferritin levels seen in our HeLa cell system and demonstrate the specific control of HFE over the Tf-mediated pathway of iron uptake. These results also have implications for the understanding of cellular iron homeostasis in organs such as the liver, pancreas, heart, and spleen that are iron loaded in hereditary hemochromatotic individuals lacking functional HFE.


Subject(s)
HLA Antigens/pharmacology , Histocompatibility Antigens Class I/pharmacology , Iron/metabolism , Membrane Proteins , Transferrin/pharmacology , Biological Transport , Endocytosis/genetics , Exocytosis/genetics , Gene Expression Regulation/drug effects , HeLa Cells , Hemochromatosis/genetics , Hemochromatosis Protein , Humans , Iodine Radioisotopes , Iron Radioisotopes , Kinetics , Nitrilotriacetic Acid/metabolism , Receptors, Transferrin/metabolism , Tetracyclines/pharmacology
10.
J Biol Chem ; 273(34): 22068-74, 1998 Aug 21.
Article in English | MEDLINE | ID: mdl-9705350

ABSTRACT

The mechanism by which a novel major histocompatibility complex class I protein, HFE, regulates iron uptake into the body is not known. HFE is the product of the gene that is mutated in >80% of hereditary hemochromatosis patients. It was recently found to coprecipitate with the transferrin receptor (Feder, J. N., Penny, D. M., Irrinki, A., Lee, V. K., Lebron, J. A., Watson, N., Tsuchihashi, Z., Sigal, E., Bjorkman, P. J., and Schatzman, R. C. (1998) Proc. Natl. Acad. Sci. U. S. A. 95, 1472-1477; Parkkila, S., Waheed, A., Britton, R. S., Bacon, B. R., Zhou, X. Y., Tomatsu, S., Fleming, R.E. , and Sly, W. S. (1997) Proc. Natl. Acad. Sci. U. S. A. 94, 13198-13202) and to decrease the affinity of transferrin for the transferrin receptor (Feder et al.). In this study, HeLa cells were transfected with HFE under the control of the tetracycline-repressible promoter. We demonstrate that HFE and the transferrin receptor are capable of associating with each other within 30 min of their synthesis with pulse-chase experiments. HFE and the transferrin receptor co-immunoprecipitate throughout the biosynthetic pathway. Excess HFE is rapidly degraded, whereas the HFE-transferrin receptor complex is stable. Immunofluorescence experiments indicate that they also endocytose into transferrin-positive compartments. Combined, these results suggest a role for the transferrin receptor in HFE trafficking. Cells expressing HFE have modestly increased levels of transferrin receptor and drastically reduced levels of ferritin. These results implicate HFE further in the modulation of iron levels in the cell.


Subject(s)
HLA Antigens/metabolism , Histocompatibility Antigens Class I/metabolism , Iron/physiology , Major Histocompatibility Complex , Membrane Proteins , Receptors, Transferrin/metabolism , Cloning, Molecular , HeLa Cells , Hemochromatosis Protein , Homeostasis , Humans , Kinetics , Promoter Regions, Genetic , Protein Synthesis Inhibitors/pharmacology , Tetracycline/pharmacology
11.
J Biol Chem ; 273(20): 12169-75, 1998 May 15.
Article in English | MEDLINE | ID: mdl-9575164

ABSTRACT

The soluble human transferrin receptor (TfR) found in blood is the result of a proteolytic cleavage occurring in the ectodomain of the receptor close to the transmembrane domain at Arg-100. We have discovered another cleavage site between Gly-91 and Val-92 even closer to the transmembrane domain. Cleavage at Gly-91 differs markedly from the normal cleavage site. It occurs when the entire cytoplasmic portion or the proximal 31 amino acids of the transmembrane domain are deleted. A soluble disulfide-bonded dimer of the TfR is released into the medium in contrast to the cleavage at Arg-100 where a dimer lacking intersubunit disulfide bonds is released. Whereas the cleavage at Arg-100 is generated by cycling through the endosomal system, pulse-chase experiments indicate that cleavage at Gly-91 occurs predominantly during the biosynthesis of the receptor. Pulse-chase analysis of the biosynthesis of mutant TfRs that lack the membrane-proximal cytoplasmic domain show that they exit the endoglycosidase H-sensitive compartment at a slower rate than the wild type TfR. These results suggest that the cytoplasmic domain influences the trafficking of the TfR either by influencing the folding of the ectodomain or by providing a positive signal for its transport through the biosynthetic pathway.


Subject(s)
Cytoplasm/metabolism , Receptors, Transferrin/metabolism , Amino Acid Sequence , Animals , Biological Transport , CHO Cells , Cell Membrane/metabolism , Cricetinae , Dimerization , Electrophoresis, Gel, Pulsed-Field , Glycine/metabolism , Hexosaminidases/metabolism , Humans , Hydrolysis , Molecular Sequence Data , Mutagenesis , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics
12.
J Biol Chem ; 273(27): 17056-63, 1998 Jul 03.
Article in English | MEDLINE | ID: mdl-9642270

ABSTRACT

Endocytosis of surface proteins through clathrin-coated pits requires an internalization signal in the cytoplasmic domain. Two types of internalization signal have been described: one requiring a tyrosine as the critical residue (tyrosine-based motif), and the other consisting of either two consecutive leucines or an isoleucine and leucine (dileucine motif). Although it seems that these signals are necessary and sufficient for endocytic targeting, the mechanism of recognition is not well understood. To examine this question, tetracycline-repressible cell lines were used to overexpress one of several receptors bearing a tyrosine-based internalization signal. By measuring the rates of endocytosis for either the overexpressed receptor, or that of other endogenous receptors, we were able to show that the endocytosis of identical receptors could be saturated, but a complete lack of competition exists between the transferrin receptor (TfR), the low-density lipoprotein receptor, and the epidermal growth factor receptor. Overexpression of any one of these receptors resulted in its redistribution toward the cell surface, implying that entry into coated pits is limited. During high levels of TfR expression, however, a significant increase in the amount of surface Lamp1, but not low-density lipoprotein receptor, epidermal growth factor receptor, or Lamp2, is detected. This suggests that Lamp1 and TfR compete for the same endocytic sites. Together, these results support the idea that there are at least three distinct saturable components involved in clathrin-mediated endocytosis.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , Receptors, LDL/metabolism , Receptors, Transferrin/metabolism , Tyrosine/metabolism , Base Sequence , Binding, Competitive , Cell Line , DNA Primers , Mutagenesis , Recombinant Proteins/metabolism
13.
Biochemistry ; 36(17): 5276-84, 1997 Apr 29.
Article in English | MEDLINE | ID: mdl-9136890

ABSTRACT

The human transferrin receptor (TfR) has three N-linked oligosaccharides. A combination of site-directed mutagenesis and carbohydrate and protein chemistry was used to characterize the structures of the N-linked oligosaccharides and to map their locations. We find that the type of oligosaccharide at each position was unique for that particular site. Human TfR isolated from placentae was used to characterize the structure of the oligosaccharides found in the native TfR. Following digestion of purified TfR with trypsin, individual peptides were obtained via RP-HPLC and were assayed for monosaccharides by strong acid hydrolysis and HPAE-PAD. Peptides containing carbohydrate were subjected to amino acid sequencing to identify the specific Asn residue. The oligosaccharides at Asn 251 are of the complex type. HPAE-PAD and FACE analysis suggests that they are triantennary and trisialylated with core fucosylation. The glycopeptide containing the site at Asn 317 was obtained after limited tryptic digestion and RP-HPLC. FACE analysis reveals predominantly a family of sialylated hybrid oligosaccharides. The consensus sequences for each N-linked site were mutated in various combinations and the resultant TfRs expressed in mouse 3T3 cells. Endoglycosidase H digestion of the mutated TfRs indicates that the pattern of oligosaccharides is consistent with the type of oligosaccharides found at each position in human tissue and the glycosylation of one site does not directly affect the glycosylation of other sites. Previous studies indicated that the oliosaccharide at Asn 727 was high-mannose type [Hayes, G. R., et al. (1995) Glycobiology 5, 227-232]. These results indicate that the type of oligosaccharide found at each site is most dependent on the environment surrounding it.


Subject(s)
Oligosaccharides/chemistry , Receptors, Transferrin/chemistry , 3T3 Cells , Animals , Carbohydrate Conformation , Carbohydrate Sequence , Carbohydrates/chemistry , Cells, Cultured , Hexosaminidases/metabolism , Humans , Mice , Models, Molecular , Molecular Sequence Data , Structure-Activity Relationship
14.
J Biol Chem ; 272(4): 2116-21, 1997 Jan 24.
Article in English | MEDLINE | ID: mdl-8999911

ABSTRACT

Cell-surface receptors that undergo clathrin-mediated endocytosis contain short amino acid sequences in their cytoplasmic domain that serve as internalization signals. Interactions between these sequences and components of the endocytic machinery should become limiting upon overexpression of the constitutively recycling transferrin receptor (TfR). A tetracycline-responsive system was used to induce overexpression of the TfR up to 20-fold in HeLa cells. Internalization assays indicate the rate of 125I-transferrin uptake per surface TfR is reduced by a factor of 4 in induced cells. Consistent with endocytosis being the rate-limiting step, TfRs shift from an endosomal to more of a plasma membrane distribution with TfR overexpression. The clathrin-associated protein AP-2 has been proposed to interact directly with the cytoplasmic domain of many receptors, yet no changes in the amount or distribution of AP-2 were detected in induced cells. The internalization rate for the epidermal growth factor receptor was also measured, with or without induction of TfR expression. Even though endocytosis of the TfR is saturated in induced cells, 125I-labeled epidermal growth factor continues to be internalized at a rate identical to that seen in uninduced cells. We propose that there are different limiting steps for the endocytosis of these two receptors.


Subject(s)
Endocytosis , ErbB Receptors/metabolism , Receptors, Transferrin/metabolism , Adaptor Protein Complex alpha Subunits , Adaptor Proteins, Vesicular Transport , Epidermal Growth Factor/metabolism , Humans , Kinetics , Membrane Proteins/metabolism , Transferrin/metabolism
15.
J Cell Biol ; 139(7): 1719-33, 1997 Dec 29.
Article in English | MEDLINE | ID: mdl-9412467

ABSTRACT

Furin catalyzes the proteolytic maturation of many proproteins within the trans-Golgi network (TGN)/endosomal system. Furin's cytosolic domain (cd) directs both the compartmentalization to and transit between its manifold processing compartments (i.e., TGN/biosynthetic pathway, cell surface, and endosomes). Here we report the identification of the first furin cd sorting protein, ABP-280 (nonmuscle filamin), an actin gelation protein. The furin cd was used as bait in a yeast two-hybrid screen to identify ABP-280 as a furin-binding protein. Binding analyses in vitro and coimmunoprecipitation studies in vivo showed that furin and ABP-280 interact directly and that ABP-280 tethers furin molecules to the cell surface. Quantitative analysis of both ABP-280-deficient and genetically replete cells showed that ABP-280 modulates the rate of internalization of furin but not of the transferrin receptor, a cycling receptor. However, although ABP-280 directs the rate of furin internalization, the efficiency of sorting of the endoprotease from the cell surface to early endosomes is independent of expression of ABP-280. By contrast, efficient sorting of furin from early endosomes to the TGN requires expression of ABP-280. In addition, ABP-280 is also required for the correct localization of late endosomes (dextran bead uptake) and lysosomes (LAMP-1 staining), demonstrating a pleiotropic role for this actin binding protein in the organization of cellular compartments and directing protein traffic. Finally, and consistent with the trafficking studies on furin, we showed that ABP-280 modulates the processing of furin substrates in the endocytic but not the biosynthetic pathways. The novel roles of ABP-280 and the cytoskeleton in the sorting of furin in the TGN/ endosomal system and the formation of proprotein processing compartments are discussed.


Subject(s)
Carrier Proteins/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational , Subtilisins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Compartmentation , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Endocytosis , Endosomes/metabolism , Filamins , Furin , Humans , Lysosomes/metabolism , Models, Biological , Molecular Sequence Data , Receptors, Transferrin/metabolism , Tumor Cells, Cultured
16.
J Cell Physiol ; 168(2): 284-93, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8707864

ABSTRACT

A soluble form of the human transferrin receptor (TfR) resulting from proteolytic cleavage at Arg 100 has been measured in human blood. In tissue culture cells elimination of the O-linked carbohydrate at Thr 104, four amino acids from the cleavage site, results in enhanced cleavage of the TfR (Rutledge et al., 1994, Blood, 83:580-586). In the present set of studies, the influence of amino acid substitution and the composition of the oligosaccharide at amino acid 104 on the cleavage of the TfR was examined. Site-directed mutagenesis was used to generate six different amino acids at position 104 which varied in size and charge. Measurement of the soluble TfR in the conditioned medium of the transfected cells of each mutant TfR showed that the large and charged side chains inhibited TfR cleavage the most. Otherwise the properties of the mutant TfRs were indistinguishable from the wild-type TfR in that the affinity of transferrin for these receptors, the extent of disulfide bond formation of the TfRs, and the proportion of TfRs at the cell surface were similar to that of the wild-type TfR. Removal of the sialic acid component of the carbohydrate from wild-type TfR by treatment of live cells with neuraminidase enhances TfR cleavage. Expression of wild-type TfR in CHO IdlD cells (a glycosylation defective cell line) also shows enhanced cleavage under conditions that produce truncated or no O-linked carbohydrates. Treatment of IdlD cells with neuraminidase reveals that the sialic acid of the O-linked carbohydrate protects against TfR cleavage, whereas the core sugars Gal-NAc and Gal do not protect as much. These results show that the terminal charged sialic acid residues are important for protection from proteolytic cleavage and suggest that cleavage could be regulated in the cell by removal of all or part of the carbohydrate.


Subject(s)
Receptors, Transferrin/metabolism , Acetylgalactosamine/metabolism , Animals , CHO Cells , Cricetinae , Electrophoresis, Polyacrylamide Gel , Endopeptidases/metabolism , Galactose/metabolism , Humans , Mutagenesis, Site-Directed , N-Acetylneuraminic Acid , Neuraminidase/metabolism , Polysaccharides/chemistry , Polysaccharides/metabolism , Precipitin Tests , Protein Binding , Protein Conformation , Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Sialic Acids/metabolism , Transferrin/metabolism
17.
Glycobiology ; 5(2): 227-32, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7780197

ABSTRACT

The human transferrin receptor (TfR) contains three N-linked oligosaccharides and glycosylation is required for the proper folding and function of the molecule. Earlier studies demonstrated that the oligosaccharide at Asn-727 is vital for the production of fully active TfR. The oligosaccharide(s) present at this site have been analysed using a combination of site-directed mutagenesis and chemical analysis. Wild-type TfR and mutants containing only the Asn-727 site or missing all three sites were transfected into mouse 3T3 cells and receptors were analysed by endo-N-acetylglucosaminidase H (Endo-H) digestion, SDS-PAGE and immunoblotting. These studies suggested that the Asn-727 site contains high-mannose or Endo-H-sensitive hybrid oligosaccharides. Glycosylation of Asn-727 found in the TfR purified from human placentae was analysed by high-pH anion-exchange chromatography with pulsed amperometric detection (HPAE-PAD) and mass spectrometry following tryptic digestion, peptide purification via reverse-phase high-performance liquid chromatography (RP-HPLC) and peptide sequencing. HPAE-PAD showed the presence of a series of high-mannose oligosaccharides. Mass spectrometry confirmed these observations, but also showed the presence of an 80 Da anionic moiety on a fraction of the oligosaccharides.


Subject(s)
Mannose/analysis , Oligosaccharides/chemistry , Receptors, Transferrin/chemistry , Receptors, Transferrin/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Asparagine/analysis , Binding Sites , Carbohydrate Sequence , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Glycosylation , Hexosaminidases , Humans , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase , Mass Spectrometry , Mice , Molecular Sequence Data , Oligosaccharides/isolation & purification , Peptide Mapping , Receptors, Transferrin/biosynthesis , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection , Trypsin
18.
J Biol Chem ; 269(50): 31864-8, 1994 Dec 16.
Article in English | MEDLINE | ID: mdl-7989360

ABSTRACT

The transmembrane protein, transferrin receptor (TfR), is found in a soluble form in human serum and in the medium of cell lines grown in tissue culture. The soluble form is generated by proteolytic cleavage between Arg-100 and Leu-101. We used two mutant human TfRs expressed in Chinese hamster ovary (CHO) cells lacking endogenous transferrin receptor to characterize the protease that cleaves the TfR and determine its location in the cell. The T104D mutant TfR lacks the O-linked carbohydrate at position 104, and is more susceptible to proteolytic cleavage at Arg-100 than the wildtype human TfR in these cells. We find that the protease is not a component of the serum in the growth medium, and it is not secreted by the cells. Cleavage does not occur during biosynthesis of the TfR, and occurs after the TfR has reached the cell surface. Expression of the T104D TfR in a temperature-sensitive acidification defective CHO cell line, G.7.1, shows that cleavage of the TfR is not dependent on acidification of endosomes. The C20A23 TfR is an endocytosis deficient mutant lacking an internalization signal. This mutant TfR, which is mainly localized to the cell surface, is cleaved less efficiently than the wild-type TfR, indicating that the protease is localized to an intracellular compartment.


Subject(s)
Endosomes/metabolism , Receptors, Transferrin/metabolism , Animals , Biological Transport , CHO Cells , Cell Compartmentation , Cricetinae , Endocytosis , Endopeptidases/metabolism , Intracellular Membranes/metabolism , Molecular Weight , Mutagenesis, Site-Directed , Recombinant Proteins , Solubility , Structure-Activity Relationship
19.
Blood ; 83(2): 580-6, 1994 Jan 15.
Article in English | MEDLINE | ID: mdl-8286753

ABSTRACT

The transferrin receptor (TfR) is the plasma membrane protein responsible for the binding and internalization of the major iron-transport protein, transferrin. The function of the single O-linked oligosaccharide near the transmembrane domain of the TfR at amino acid Thr 104 is unknown. To elucidate the effect of the O-linked carbohydrate on TfR function, the oligosaccharide was eliminated by replacing Thr 104 with Asp and the mutated cDNA was expressed in a cell line lacking endogenous TfR. Elimination of the oligosaccharide at Thr 104 results in a form of the receptor that is susceptible to cleavage. A 78-kD soluble TfR that can bind transferrin is released into the growth medium. The intact mutant TfR is not grossly altered in its structure and does not differ significantly from the wild-type human receptor in many respects: (1) It shows the same distribution between the plasma membrane and intracellular compartments; (2) the binding constant for transferrin is similar to that of the wild-type TfR; and (3) it is not rapidly degraded. Protein-sequence analysis of the soluble form indicates that the sequence begins at amino acid 101 of the intact receptor. This is the same cleavage site reported for a soluble form of normal receptor found in human serum. Substitution of Gly, Glu, or Met at position 104 also results in increased cleavage of the TfR and suggests that elimination of the O-linked carbohydrate at position 104 enhances the susceptibility of TfR to cleavage and may mimic a naturally occurring process previously described as being related to erythropoiesis.


Subject(s)
Receptors, Transferrin/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Erythropoiesis , Glycosylation , Humans , Molecular Sequence Data , Mutation , Receptors, Transferrin/chemistry , Threonine
20.
J Biol Chem ; 268(17): 12780-6, 1993 Jun 15.
Article in English | MEDLINE | ID: mdl-8509412

ABSTRACT

The transferrin receptor is a cell surface protein and is responsible for the uptake of iron into many eukaryotic cells. In its mature form, the receptor possesses three asparagine-linked oligosaccharides. The effect of asparagine-linked glycosylation on the processing and cell surface localization of the human transferrin receptor is examined here by site-directed mutagenesis. Each of the extracellular consensus sequences (Asn-X-Ser/Thr) for asparagine-linked glycosylation was mutated individually and in all possible combinations. The constructs were transfected stably into NIH-3T3 cells and a Chinese hamster ovary cell line lacking endogenous transferrin receptors. Of the seven possible combinations of glycosylation sites, single mutations eliminating glycosylation at either Asn251 or Asn317 do not affect the processing and surface localization of the receptor. Eliminating both of these sites together has a small effect on the behavior of the receptor. However, mutation of the C-terminal glycosylation site (Asn727) has the most profound negative effect on the appearance of the receptor at the cell surface. The mutants lacking glycosylation at Asn727 appear to be retained in the endoplasmic reticulum as an increased association with binding immunoglobulin protein (BiP) is observed. Addition of a new glycosylation site in the C-terminal region of the unglycosylated mutated transferrin receptor restores the cell surface localization and the transferrin binding of the transferrin receptor, indicating that glycosylation in this region is critical for the correct transport of this receptor to the cell surface.


Subject(s)
Protein Processing, Post-Translational , Receptors, Transferrin/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Membrane/metabolism , DNA , DNA Mutational Analysis , Glycosylation , Humans , Kinetics , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligodeoxyribonucleotides , Receptors, Transferrin/isolation & purification , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL