Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Diabetes Obes Metab ; 20(9): 2075-2084, 2018 09.
Article in English | MEDLINE | ID: mdl-29749030

ABSTRACT

We describe a newly identified therapeutic target for type 1 diabetes (T1D): an envelope protein of endogenous retroviral origin, human endogenous retrovirus W envelope (HERV-W-Env). HERV-W-Env was found to be detected in the blood of ~60% of patients with T1D and is expressed in acinar pancreatic cells of 75% of patients with T1D at post mortem examination. Preclinical experiments showed that this protein displays direct cytotoxicity on human ß-islet cells. In vivo HERV-W-Env impairs the insulin and glucose metabolism in transgenic mice expressing HERV-W-Env. GNbAC1, an IgG4 monoclonal antibody, has been developed to specifically target HERV-W-Env and to neutralize the effect of HERV-W-Env in vitro and in vivo. GNbAC1 is currently in clinical development for multiple sclerosis and > 300 subjects have been administered with GNbAC1 so far. GNbAC1 is now being tested in T1D in the RAINBOW-T1D study, which is a randomized placebo-controlled study with the objective of showing the safety and pharmacodynamic response of GNbAC1 in patients who have had T1D with a maximum of 4 years' duration. GNbAC1 is being tested vs placebo at the dose of 6 mg/kg in 60 patients during six repeated administrations for 6 months; a 6-month open-label extension will follow. The primary endpoint is to assess safety, and secondary endpoints are the pharmacodynamic responses to GNbAC1. GNbAC1 targeting HERV-W-Env is currently in clinical development in T1D, with the first safety and pharmacodynamic study. If the study results are positive, this may open the door to the development of an innovative non-immunomodulatory disease-modifying treatment for T1D.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Endogenous Retroviruses/drug effects , Gene Products, env/drug effects , Immunologic Factors/pharmacology , Diabetes Mellitus, Type 1/virology , Endogenous Retroviruses/immunology , Gene Products, env/blood , Gene Products, env/immunology , Humans
2.
Mutagenesis ; 31(6): 695-702, 2016 11.
Article in English | MEDLINE | ID: mdl-27576335

ABSTRACT

Human endogenous retrovirus (HERV) sequences make up ~8% of the human genome and increased expression of some HERV proteins has been observed in various pathologies including leukaemia and multiple sclerosis. However, little is known about the function of these HERV proteins or environmental factors which regulate their expression. Silver nanoparticles (AgNPs) are used very extensively as antimicrobials and antivirals in numerous consumer products although their effect on the expression of HERV gene products is unknown. Cell proliferation and cell toxicity assays were carried out on human acute T lymphoblastic leukaemia (MOLT-4) and Fanconi anaemia associated acute myeloid leukaemia (FA-AML1) cells treated with two different sizes of AgNPs (7nm and 50nm diameter). Reverse-transcriptase polymerase chain reaction and western blotting were then used to the assess expression of HERV-W syncytin-1 mRNA and protein in these cells. FA-AML1 cells were more sensitive overall than MOLT-4 to treatment with the smaller 7nm sized AgNp's being the most toxic in these cells. MOLT-4 cell were more resistant and showed no evidence of differential toxicity to the different sized particles. Syncytin-1 mRNA and protein were induced by both 7 and 50nm AgNPs in both cell types yet with different kinetics. In summary, the observation that AgNPs induce expression of syncytin-1 in FA-AML1 and MOLT-4 cells at doses as little as 5 µg/ml is grounds for concern since this protein is up-regulated in both malignant and neurodegenerative diseases. Considering the widespread use of AgNPs in the environment it is clear that their ability to induce syncytin-1 should be investigated further in other cell types.


Subject(s)
Gene Products, env/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, T-Cell/drug therapy , Metal Nanoparticles/toxicity , Pregnancy Proteins/drug effects , Silver/toxicity , Up-Regulation , Cell Proliferation , Endogenous Retroviruses/metabolism , Fanconi Anemia/complications , Gene Expression Regulation, Leukemic , Gene Products, env/genetics , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/physiopathology , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/physiopathology , Metal Nanoparticles/chemistry , Pregnancy Proteins/genetics , RNA, Messenger , Silver/pharmacology
3.
Cell Biol Int ; 33(1): 36-48, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18992834

ABSTRACT

Exosomes are small membrane vesicles that are released into the extracellular compartment as a consequence of fusion of multivesicular endosomes with the plasma membrane. To unravel the molecular basis of protein sorting into exosomes, we have made a chimeric protein containing the cytosolic domain of the transmembrane subunit of the viral Env protein of BLV and the ectodomain of CD8 (CDTM-BLV-CD8). When expressed in K562 cells known to constitutively secrete exosomes, the chimera was found to be very efficiently targeted to the released vesicles. Very interestingly, the cytosolic domain of the Env protein contains peptide motifs potentially recognized by components of the ESCRT machinery that could be related to chimera sorting into the vesicles. Then, quantifying the chimera secretion, we investigated the site of exosome biogenesis in K562 cells using a pharmacological approach. We present different arguments indicating that CDTM-BLV-CD8-containing exosomes are likely formed from a recycling endosomal/TGN compartment.


Subject(s)
Exocytosis , Exosomes/metabolism , Gene Products, env/metabolism , Leukemia Virus, Bovine , Amino Acid Motifs , Amino Acid Sequence , Animals , Cattle , Cell Line, Tumor , Cell Membrane/physiology , Fluorescent Antibody Technique, Direct , Gene Products, env/drug effects , Gene Products, env/genetics , Humans , K562 Cells , Molecular Sequence Data , Protein Transport , Rats , Receptors, Antigen, T-Cell/drug effects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , trans-Golgi Network/metabolism
4.
Antimicrob Agents Chemother ; 53(2): 678-87, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19047654

ABSTRACT

The human T-cell lymphotropic virus type 1 (HTLV-1) is the cause of adult T-cell leukemia and inflammatory diseases including HTLV-1-associated myelopathy/tropical spastic paraparesis. HTLV-1 can be transmitted through sexual contact, mother-to-child transmission, and exposure to contaminated blood. Microbicides are agents that interfere with microbial infectivity at mucous membranes, and candidates are under development for use against sexually transmitted viruses such as human immunodeficiency virus type 1. We previously demonstrated that cell surface polyanionic heparan sulfate proteoglycans bind the HTLV-1 envelope glycoprotein surface subunit gp46, facilitating cell-cell and cell-free virus spread in vitro. We now show, using assays for Env-receptor binding inhibition, Env-induced cell-cell fusion, cell-cell virus spread, and pseudotype HTLV-1 infectivity, that the soluble polyanions PRO 2000 and dextran sulfate are potent inhibitors of HTLV-1 spread in vitro, with PRO 2000 being the more promising candidate. The results of these studies suggest that candidate topical microbicides may be of use in reducing HTLV-1 sexual transmission.


Subject(s)
Antiviral Agents/pharmacology , HTLV-I Infections/prevention & control , Human T-lymphotropic virus 1/drug effects , Naphthalenesulfonates/pharmacology , Polymers/pharmacology , Antiviral Agents/chemistry , Cell Fusion , Cell Survival/drug effects , Cells, Cultured , Gene Products, env/drug effects , HTLV-I Infections/transmission , HeLa Cells , Humans , Naphthalenesulfonates/chemistry , Peptides/pharmacology , Polymers/chemistry , Retroviridae Proteins, Oncogenic/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Virion/drug effects
5.
Curr HIV Res ; 5(2): 235-50, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17346137

ABSTRACT

BACKGROUND: Highly active antiretroviral therapy (HAART) can successfully reduce plasma and tissue levels of HIV-1 RNA and results in reductions in HIV-related morbidity and mortality, but the slow viral evolution during therapy in cellular reservoirs is a continuing problem. In addition, little remains known how viral evolutionary process may differ between cell-free and cell-associated compartments, over time, in vivo in patients receiving HAART or STI. OBJECTIVES: The main objectives of this study were to assess viral replication kinetics, drug resistance and viral evolution during HAART and STI. STUDY DESIGN: We have conducted a longitudinal study of virus culture kinetics in vitro, molecular analysis of uncultured HIV-1 variants from plasma and PBMC of 6 patients on HAART, 4 patients on STI, and 6 from treatment-naïve patients. RESULTS: Our data suggest that drug resistance mutations remained compartmentalized between plasma and PBMC. The divergent distribution of resistance mutations between plasma and PBMC coincided with divergent env gene evolution in these compartments. In contrast, the HIV strains from therapy-naive patients showed tight genetic and phylogenetic concordance between plasma and PBMC. Both STI and non-STI groups showed the presence of resistance mutations to both RT and protease inhibitors, which correlated with inadequate suppression of viremia and partially with the virus culture isolation in vitro. CONCLUSIONS: Overall, STI for HIV patients has no added advantage over regular HAART at the virologic level and in the diminution of resistance mutations that result in therapy failure. Under both forms of anti-retroviral therapies, virus could be isolated in vitro from the PBMC showing continuing low-level viral replication under suppressive therapy. Overall, these data may be useful in predicting the late emergence of drug resistance mutations via the latent integrated provirus.


Subject(s)
Antiretroviral Therapy, Highly Active/methods , Drug Resistance, Viral/genetics , Evolution, Molecular , HIV-1/drug effects , Leukocytes, Mononuclear/virology , Plasma/virology , Virus Replication/drug effects , Adult , Cell Culture Techniques , Drug Administration Schedule , Gene Products, env/drug effects , Gene Products, env/genetics , Genes, env/drug effects , HIV Protease/drug effects , HIV Protease/genetics , HIV Reverse Transcriptase/drug effects , HIV Reverse Transcriptase/genetics , Humans , Male , Middle Aged , Molecular Sequence Data , Phylogeny , Sequence Analysis, Protein , Viral Load , Virus Replication/genetics
7.
Proc Natl Acad Sci U S A ; 95(24): 14435-40, 1998 Nov 24.
Article in English | MEDLINE | ID: mdl-9826718

ABSTRACT

Previously, we showed that the addition of human erythrocyte glycosphingolipids (GSLs) to nonhuman CD4(+) or GSL-depleted human CD4(+) cells rendered those cells susceptible to HIV-1 envelope glycoprotein-mediated cell fusion. Individual components in the GSL mixture were isolated by fractionation on a silica-gel column and incorporated into the membranes of CD4(+) cells. GSL-supplemented target cells were then examined for their ability to fuse with TF228 cells expressing HIV-1LAI envelope glycoprotein. We found that one GSL fraction, fraction 3, exhibited the highest recovery of fusion after incorporation into CD4(+) nonhuman and GSL-depleted HeLa-CD4 cells and that fraction 3 contained a single GSL fraction. Fraction 3 was characterized by MS, NMR spectroscopy, enzymatic analysis, and immunostaining with an antiglobotriaosylceramide (Gb3) antibody and was found to be Gal(alpha1-->4)Gal(beta1-->4)Glc-Cer (Gb3). The addition of fraction 3 or Gb3 to GSL-depleted HeLa-CD4 cells recovered fusion, but the addition of galactosylceramide, glucosylceramide, the monosialoganglioside, GM3, lactosylceramide, globoside, the disialoganglioside, GD3, or alpha-galactosidase A-digested fraction 3 had no effect. Our findings show that the neutral GSL, Gb3, is required for CD4/CXCR4-dependent HIV-1 fusion.


Subject(s)
CD4 Antigens/physiology , Cell Fusion/physiology , Gene Products, env/physiology , HIV-1/physiology , Receptors, CXCR4/physiology , Trihexosylceramides/pharmacology , Acetylation , Carbohydrate Conformation , Carbohydrate Sequence , Cell Fusion/drug effects , Cell Line , Erythrocytes/chemistry , Erythrocytes/physiology , Gene Products, env/drug effects , HIV-1/drug effects , HeLa Cells , Humans , Mass Spectrometry , Molecular Conformation , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Receptors, CXCR4/drug effects , Trihexosylceramides/blood , Trihexosylceramides/chemistry
8.
J Photochem Photobiol B ; 44(1): 77-83, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9745730

ABSTRACT

The photodynamic inactivation of herpes simplex virus type 1 (HSV-1) by two phthalocyanines (Pcs), the cationic dye HOSi-PcOSi(CH3)2(CH2)3N+(CH3)3I-(Pc5) and the amphiphilic dye aluminum dibenzodisulfophthalocyanine hydroxide (AlN2SB2POH), has been compared with that by the anionic dye, Merocyanine 540 (Mc540). Both Pc derivatives demonstrate a remarkable virucidal activity upon light activation even 3 h after the onset of HSV-1 adsorption, while Mc540 is effective for only 30 min after adsorption. Since fusion and virus penetration are promoted by membrane glycoproteins, we have studied the damage to viral proteins following photodynamic treatment (PDT) of HSV-1 and its relation to inactivation. The effect of AlN2SB2POH PDT is assessed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). Major changes are found in the protein profile of PDT-treated HSV-1. A reduced ability of specific antibodies to react with HSV-1 major envelope proteins is detected by employing the Western blot assay. In particular, we demonstrate the related changes of glycoprotein D (gD), a structural protein of the HSV envelope. Since the envelope proteins participate in viral entry into the host cell, these alterations to viral envelope proteins may impair their ability to participate in early events of viral entry, leading to reduced infectivity of HSV-1. In contrast, no significant changes in the proteins' electrophoretic mobility could be seen after PDT with Mc540 or with Pc5. When HSV-1 purified proteins are subjected to combined electrophoretic and electro osmotic forces on cellulose acetate, there is a shift in their cathode mobility, which may indicate changes in the protein mass and protein net charges following AlN2SB2POH photosensitization. There are only minor changes in the virus proteins, assayed as above, when HSV-1 is treated with Pc5.


Subject(s)
Gene Products, env/drug effects , Indoles/pharmacology , Photosensitizing Agents/pharmacology , Simplexvirus/drug effects , Viral Proteins/drug effects , Viral Proteins/radiation effects , Animals , Chlorocebus aethiops , Gene Products, env/radiation effects , Isoindoles , Kinetics , Light , Pyrimidinones/pharmacology , Simplexvirus/physiology , Simplexvirus/radiation effects , Vero Cells
9.
Proc Natl Acad Sci U S A ; 95(20): 11511-3, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751696

ABSTRACT

The success of highly active anti-retroviral therapy (HAART) has inspired new concepts for eliminating HIV from infected individuals. A major obstacle is the persistence of long-lived reservoirs of latently infected cells that might become activated at some time after cessation of therapy. We propose that, in the context of treatment strategies to deliberately activate and eliminate these reservoirs, hybrid toxins targeted to kill HIV-infected cells be reconsidered in combination with HAART. Such combinations might also prove valuable in protocols aimed at preventing mother-to-child transmission and establishment of infection immediately after exposure to HIV. We suggest experimental approaches in vitro and in animal models to test various issues related to safety and efficacy of this concept.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Toxins, Biological/therapeutic use , Animals , Clinical Trials, Phase I as Topic , Exotoxins/adverse effects , Exotoxins/therapeutic use , Female , Gene Products, env/drug effects , HIV/drug effects , HIV Infections/transmission , HIV Infections/virology , Humans , Immunotoxins/adverse effects , Immunotoxins/therapeutic use , Infectious Disease Transmission, Vertical , Liver/drug effects , Models, Biological , Pregnancy , Proviruses/drug effects , Recombinant Proteins/adverse effects , Recombinant Proteins/therapeutic use , Virus Activation/drug effects
10.
Virology ; 247(2): 137-43, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9705906

ABSTRACT

A 22-amino-acid-long multibranched peptide construct (CLV) derived from the cleavage region (KIEPLGVAPTKAKRR*VVQREKR*) of the human immunodeficiency virus (HIV) type-1 envelope precursor inhibits HIV infection (Virology, 1996, 223, 406-408). We attempted to characterize its activity for Env expressed via a recombinant vaccinia virus (rVV): gp 160 cleavage was delayed, but not impaired, in the presence of CLV (10 microM), whereas neither Env production nor Env membrane expression was significantly altered. Through the synthesis of analogs, we concluded that the presence of a cleavage sequence was required for inhibition of syncytium formation by CLV in rVV-infected CD(4+) cell cultures: indeed, a single amino acid residue substitution (R* > S) in the cleavage sites presented by CLV abolished its activity. Other analogs allowed us to further determine the region of CLV which mediates its activity. The ability of a radiolabeled CLV analog to enter cells was also shown. Although, these data strongly suggest that CLV acts on Env fusogenicity at least partially through interference with gp160 processing.


Subject(s)
Anti-HIV Agents/pharmacology , Gene Products, env/metabolism , HIV Envelope Protein gp160/metabolism , Peptides/pharmacology , Amino Acid Sequence , Animals , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/metabolism , Cell Line , Cell Membrane/virology , Cells, Cultured , Cricetinae , Gene Products, env/drug effects , Giant Cells/virology , HIV Envelope Protein gp160/drug effects , Humans , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/metabolism , Protein Precursors/chemistry , Protein Precursors/pharmacology , Protein Processing, Post-Translational/drug effects
11.
J Virol ; 72(4): 3394-400, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9525669

ABSTRACT

Monocytes/macrophages (M/M) and CD4+ T cells are two important targets of human immunodeficiency virus (HIV) infection. Different strains of HIV-1 vary markedly in their abilities to infect cells belonging to the M/M lineage. Macrophagetropic (M-tropic) HIV-1 strains replicate well in primary lymphocytes as well as in primary macrophages; however, they generally infect T-cell lines poorly, if at all. Although promonocytic cell lines such as U937 have been used as in vitro models of HIV-1 infection of M/M, these cell lines are susceptible to certain T-cell-tropic (T-tropic) HIV-1 strains but are resistant to M-tropic HIV-1. In this study, we demonstrate that (i) certain U937 clones ("plus" clones), which are susceptible only to T-tropic HIV-1, become highly susceptible to M-tropic HIV-1 upon differentiation with retinoic acid (RA); (ii) other U937 clones ("minus" clones), which are resistant to both T- and M-tropic HIV-1, remain resistant to both viruses; and (iii) RA treatment induces expression of CCR5, a fusion/entry cofactor for M-tropic HIV-1 in both types of U937 clones, and yet enhances the fusogenicity of the plus clones, but not the minus clones, with M-tropic Env's. These results indicate that the major restriction of M-tropic HIV-1 infection in promonocytic cells occurs at the fusion/entry level, that differentiation into macrophage-like phenotypes renders some of these cells highly susceptible to infection with M-tropic HIV-1, and that CD4 and CCR5 may not be the only determinants of fusion/entry of M-tropic HIV-1 in these cells.


Subject(s)
HIV-1/metabolism , Macrophages/virology , Membrane Fusion , Monocytes/virology , Cell Differentiation , Gene Products, env/drug effects , Gene Products, env/metabolism , HIV-1/drug effects , HIV-1/physiology , Humans , Ligands , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , Phenotype , Receptors, CCR5/biosynthesis , Tretinoin/pharmacology , Tumor Cells, Cultured
12.
Clin Perinatol ; 21(1): 39-73, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8013186

ABSTRACT

In the past 10 years, a large number of investigators have produced an enormous amount of information concerning the molecular biology of HIV. These studies at the most basic biological level have provided essential insights into the pathogenesis of the disease. They have supplied the information necessary for the creation of the antiviral therapies now available and have indicated the direction for the development of new therapies now in clinical trials and under investigation. Although the relatively ineffective therapies currently available serve as a constant source of disappointment for those practitioners who care for HIV-infected patients, there is some comfort to be gained from the rapid pace of investigation into the basic biology of the virus and the certainty that any more effective therapy must build upon the basic biological knowledge already obtained. A detailed study of some of the unique features observed during pediatric and perinatal HIV infection, particularly the relatively shortened time from infection to symptoms and the relative importance of CNS disease, may suggest new therapeutic approaches that will benefit both adult and pediatric patients. Finally, a comprehensive knowledge of HIV biology is an essential requirement for therapeutic maneuvers designed to interrupt the transmission of HIV from mother to child.


Subject(s)
DNA, Viral , Gene Products, env , Gene Products, gag , Gene Products, pol , HIV , Viral Regulatory and Accessory Proteins , Female , Gene Expression Regulation, Viral , Gene Products, env/antagonists & inhibitors , Gene Products, env/chemistry , Gene Products, env/drug effects , Gene Products, env/genetics , Gene Products, env/ultrastructure , Gene Products, gag/antagonists & inhibitors , Gene Products, gag/chemistry , Gene Products, gag/drug effects , Gene Products, gag/genetics , Gene Products, gag/ultrastructure , Gene Products, pol/antagonists & inhibitors , Gene Products, pol/chemistry , Gene Products, pol/drug effects , Gene Products, pol/genetics , Gene Products, pol/ultrastructure , HIV/chemistry , HIV/genetics , HIV/growth & development , HIV/physiology , HIV/ultrastructure , HIV Infections/congenital , HIV Infections/microbiology , HIV Infections/prevention & control , HIV Infections/transmission , Humans , Infant, Newborn , Molecular Biology , Pregnancy , Pregnancy Complications, Infectious/microbiology , Pregnancy Complications, Infectious/prevention & control , Regulatory Sequences, Nucleic Acid , Time Factors , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/ultrastructure , Transcription, Genetic , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/drug effects , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/ultrastructure , Virion/chemistry , Virion/genetics , Virion/growth & development , Virion/physiology , Virion/ultrastructure , Virus Integration , Virus Replication
13.
J Virol ; 68(1): 463-8, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8254757

ABSTRACT

The human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein has been shown to be extensively modified by N-linked glycosylation; however, the presence of O-linked carbohydrates on the glycoprotein has not been firmly established. We have found that enzymatic deglycosylation of the HIV-1 envelope glycoprotein with neuraminidase and O-glycosidase results in a decrease in the apparent molecular weight of the envelope glycoprotein. This result was observed in both vaccinia virus recombinant-derived envelope glycoproteins and glycoproteins derived from the IIIB, SG3, and HXB2, strains of HIV-1. The decrease in molecular weight was also observed when the envelope glycoprotein had been deglycosylated with N-glycanase F after treatment with neuraminidase and O-glycosidase, indicating that the decrease in apparent molecular weight was not attributable to the removal of N-linked carbohydrate. Treatment with neuraminidase, O-glycosidase, and N-glycanase F was found to be necessary to remove all radiolabel from [3H]glucosamine-labelled envelope glycoprotein, a result seen for both recombinant and HIV-1-derived envelope glycoprotein. [3H]glucosamine-labelled carbohydrates liberated by O-glycosidase treatment were separated by paper chromatography and were found to be of a size consistent with O-linked oligosaccharides. We, therefore, conclude that the HIV-1 envelope glycoprotein is modified by the addition of O-linked carbohydrates.


Subject(s)
Gene Products, env/chemistry , Glycoproteins/chemistry , HIV-1/chemistry , Oligosaccharides/chemistry , Amidohydrolases/pharmacology , Animals , Cells, Cultured , Chromatography, Paper , Gene Products, env/drug effects , Gene Products, env/genetics , Glucosamine/metabolism , Glycoproteins/drug effects , Glycoproteins/genetics , Glycosylation , Humans , Mannosyl-Glycoprotein Endo-beta-N-Acetylglucosaminidase/pharmacology , Molecular Weight , Neuraminidase/pharmacology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Polysaccharides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/drug effects
14.
Virology ; 197(2): 549-57, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8249278

ABSTRACT

Binding of viral envelope proteins to specific receptors on human hepatocytes is considered to be an important step in HBV infection. In this study, we demonstrate that a 34-kDa human liver plasma membrane protein specifically binds to small HBsAg in a Ca(2+)-dependent manner. By partial amino acid sequence analysis of preparatively isolated 34-kDa protein comigrating with HBsAg-binding protein obtained from binding assay on IEF/SDS-PAGE, we have identified this HBsAg-binding protein as Endonexin II (E-II). Native human liver E-II inhibits binding of HBsAg to intact human hepatocytes and shows specific binding to small HBsAg. This binding can be inhibited by human liver plasma membrane proteins, recombinant E-II, or anti-E-II antibodies. Despite 90% sequence homology, rat liver E-II does not bind to small HBsAg and does not inhibit significantly (less than 20%) binding of HBsAg to intact hepatocytes. Cross-linking of small HBsAg and radiolabeled human liver E-II resulted in a specific additional protein complex on PAGE with an apparent molecular weight of 90 kDa, corresponding to a complex of E-II and small HBsAg with a ratio of 2 to 1 or 1 to 2. These findings indicate that E-II, found in human liver, is a specific HBsAg-binding protein and might play an important role in the initiation of HBV infection.


Subject(s)
Annexin A5/metabolism , Gene Products, env/metabolism , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/metabolism , Liver/metabolism , Receptors, Virus/metabolism , Amino Acid Sequence , Animals , Annexin A5/isolation & purification , Annexin A5/pharmacology , Calcium/pharmacology , Cell Membrane/metabolism , Cross-Linking Reagents , Gene Products, env/drug effects , Hepatitis B Surface Antigens/drug effects , Humans , Molecular Sequence Data , Rats , Receptors, Virus/drug effects , Receptors, Virus/isolation & purification , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Sequence Analysis
15.
AIDS Res Hum Retroviruses ; 7(8): 707-12, 1991 Aug.
Article in English | MEDLINE | ID: mdl-1718346

ABSTRACT

The processing and secretion of the envelope glycoproteins of human immunodeficiency virus type 1 (HIV-1) were studied in chronically infected T cells and in primary macrophages treated with an antiviral antibiotic brefeldin A (BFA). BFA blocks the egress of proteins from the endoplasmic reticulum and has a profound effect on the structure of cis/medial Golgi. In MOLT-3 cells infected with the IIIB strain of HIV-1 (MOLT-3/IIIB), BFA inhibited the intracellular processing of gp160. The secretion of envelope proteins from these cells was significantly inhibited in the presence of BFA. The gag proteins, on the other hand, were processed and secreted normally. BFA also inhibited the proteolytic processing of gp160 in primary macrophages infected with HIV-1. The infectivity of virus pelleted from the medium of MOLT-3/IIIB cells treated with BFA was markedly lower than that obtained from untreated cells. These results demonstrate that the proteolytic processing of gp160 in HIV-1-infected cells takes place after the glycoprotein exists the endoplasmic reticulum and that the transport of glycoprotein to the cell surface is required for assembly of complete HIV-1 particles.


Subject(s)
Antiviral Agents/pharmacology , Cyclopentanes/pharmacology , Gene Products, env/drug effects , HIV-1/drug effects , Protein Precursors/drug effects , Protein Processing, Post-Translational/drug effects , Brefeldin A , Carbohydrate Sequence , Cell Fractionation , Cell Line , Gene Products, env/metabolism , HIV Envelope Protein gp160 , HIV Reverse Transcriptase , HIV Seropositivity/microbiology , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Macrophages/microbiology , Molecular Sequence Data , Precipitin Tests , Protein Precursors/metabolism , RNA-Directed DNA Polymerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...