Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
Nucleic Acids Res ; 50(D1): D934-D942, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34634807

ABSTRACT

Viral infectious diseases are a devastating and continuing threat to human and animal health. Receptor binding is the key step for viral entry into host cells. Therefore, recognizing viral receptors is fundamental for understanding the potential tissue tropism or host range of these pathogens. The rapid advancement of single-cell RNA sequencing (scRNA-seq) technology has paved the way for studying the expression of viral receptors in different tissues of animal species at single-cell resolution, resulting in huge scRNA-seq datasets. However, effectively integrating or sharing these datasets among the research community is challenging, especially for laboratory scientists. In this study, we manually curated up-to-date datasets generated in animal scRNA-seq studies, analyzed them using a unified processing pipeline, and comprehensively annotated 107 viral receptors in 142 viruses and obtained accurate expression signatures in 2 100 962 cells from 47 animal species. Thus, the VThunter database provides a user-friendly interface for the research community to explore the expression signatures of viral receptors. VThunter offers an informative and convenient resource for scientists to better understand the interactions between viral receptors and animal viruses and to assess viral pathogenesis and transmission in species. Database URL: https://db.cngb.org/VThunter/.


Subject(s)
Databases, Factual , Genome, Viral , Host-Pathogen Interactions/genetics , Receptors, Virus/genetics , Software , Virus Diseases/genetics , Viruses/genetics , Animals , Binding Sites , Datasets as Topic , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Internet , Molecular Sequence Annotation , Protein Binding , Receptors, Virus/classification , Receptors, Virus/metabolism , Signal Transduction , Single-Cell Analysis , Virus Diseases/metabolism , Virus Diseases/transmission , Virus Diseases/virology , Viruses/classification , Viruses/metabolism , Viruses/pathogenicity
2.
Arch Virol ; 166(7): 1811-1817, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33778918

ABSTRACT

Coronaviruses are a large family of important pathogens that cause human and animal diseases. At the end of 2019, a pneumonia epidemic caused by a novel coronavirus brought attention to coronaviruses. Exploring the interaction between the virus and its receptor will be helpful in developing preventive vaccines and therapeutic drugs. The coronavirus spike protein (S) plays an important role in both binding to receptors on host cells and fusion of the viral membrane with the host cell membrane. This review introduces the structure and function of the S protein and its receptor, focusing on the binding mode and binding region of both.


Subject(s)
Coronavirus/metabolism , Receptors, Virus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Binding Sites , COVID-19/metabolism , COVID-19/virology , Coronavirus/chemistry , Coronavirus/physiology , Humans , Protein Binding , Protein Conformation , Receptors, Virus/classification , SARS-CoV-2/chemistry , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/classification , Virus Internalization
3.
Comb Chem High Throughput Screen ; 24(10): 1769-1783, 2021.
Article in English | MEDLINE | ID: mdl-33172369

ABSTRACT

BACKGROUND: Novel coronavirus SARS-CoV-2 is responsible for the COVID-19 pandemic. It was first reported in Wuhan, China, in December 2019, and despite the tremendous efforts to control the disease, it has now spread almost all over the world. The interaction of SARSCoV- 2spike protein and its acceptor protein ACE2 is an important issue in determining viral host range and cross-species infection, while the binding capacity of spike protein to ACE2 of different species is unknown. OBJECTIVE: The present study has been conducted to determine the susceptibility of livestock, poultry and pets to SARS-CoV-2. METHODS: We evaluated the receptor-utilizing capability of ACE2s from various species by sequence alignment, phylogenetic clustering and protein-ligand interaction studies with the currently known ACE2s utilized by SARS-CoV-2. RESULT: In-silico study predicted that SARS-CoV-2 tends to utilize ACE2s of various animal species with varied possible interactions. The probability of the receptor utilization will be greater in horse and poor in chicken, followed by ruminants. CONCLUSION: Present study predicted that SARS-CoV-2 tends to utilize ACE2s of various livestock and poultry species with greater probability in equine and poor in chicken. The study may provide important insights into the animal models for SARS-CoV-2 and animal management for COVID- 19 control.


Subject(s)
Angiotensin-Converting Enzyme 2/genetics , COVID-19/epidemiology , COVID-19/virology , Pandemics , Receptors, Virus/genetics , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/classification , Angiotensin-Converting Enzyme 2/metabolism , Animals , Binding Sites , Buffaloes , COVID-19/transmission , Camelus , Cats , Cattle , Chickens , Chiroptera , Dogs , Gene Expression , Goats , Horses , Humans , Molecular Docking Simulation , Phylogeny , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Virus/chemistry , Receptors, Virus/classification , Receptors, Virus/metabolism , SARS-CoV-2/pathogenicity , Sheep , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Swine
4.
Eur Respir J ; 56(3)2020 09.
Article in English | MEDLINE | ID: mdl-32675206

ABSTRACT

In December 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged, causing the coronavirus disease 2019 (COVID-19) pandemic. SARS-CoV, the agent responsible for the 2003 SARS outbreak, utilises angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2) host molecules for viral entry. ACE2 and TMPRSS2 have recently been implicated in SARS-CoV-2 viral infection. Additional host molecules including ADAM17, cathepsin L, CD147 and GRP78 may also function as receptors for SARS-CoV-2.To determine the expression and in situ localisation of candidate SARS-CoV-2 receptors in the respiratory mucosa, we analysed gene expression datasets from airway epithelial cells of 515 healthy subjects, gene promoter activity analysis using the FANTOM5 dataset containing 120 distinct sample types, single cell RNA sequencing (scRNAseq) of 10 healthy subjects, proteomic datasets, immunoblots on multiple airway epithelial cell types, and immunohistochemistry on 98 human lung samples.We demonstrate absent to low ACE2 promoter activity in a variety of lung epithelial cell samples and low ACE2 gene expression in both microarray and scRNAseq datasets of epithelial cell populations. Consistent with gene expression, rare ACE2 protein expression was observed in the airway epithelium and alveoli of human lung, confirmed with proteomics. We present confirmatory evidence for the presence of TMPRSS2, CD147 and GRP78 protein in vitro in airway epithelial cells and confirm broad in situ protein expression of CD147 and GRP78 in the respiratory mucosa.Collectively, our data suggest the presence of a mechanism dynamically regulating ACE2 expression in human lung, perhaps in periods of SARS-CoV-2 infection, and also suggest that alternative receptors for SARS-CoV-2 exist to facilitate initial host cell infection.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections , Pandemics , Peptidyl-Dipeptidase A , Pneumonia, Viral , Serine Endopeptidases , Angiotensin-Converting Enzyme 2 , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/virology , Endoplasmic Reticulum Chaperone BiP , Gene Expression , Gene Expression Profiling/methods , Humans , Lung/metabolism , Lung/virology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/virology , Receptors, Virus/classification , Receptors, Virus/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , SARS-CoV-2 , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Virus Internalization
5.
J Med Virol ; 92(10): 2105-2113, 2020 10.
Article in English | MEDLINE | ID: mdl-32383269

ABSTRACT

Coronavirus disease-2019 (COVID-19) outbreak due to novel coronavirus or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has come out as a major threat for mankind in recent times. It is continually taking an enormous toll on mankind by means of increasing number of deaths, associated comorbidities, and socioeconomic loss around the globe. Unavailability of chemotherapeutics/vaccine has posed tremendous challenges to scientists and doctors for developing an urgent therapeutic strategy. In this connection, the present in silico study aims to understand the sequence divergence of spike protein (the major infective protein of SARS-CoV-2), its mode of interaction with the angiotensin-converting enzyme-2 receptor (ACE2) receptor of human and related animal hosts/reservoir. Moreover, the involvement of the human Toll-like receptors (TLRs) against the spike protein has also been demonstrated. Our data indicated that the spike glycoprotein of SARS-CoV-2 is phylogenetically close to bat coronavirus and strongly binds with ACE2 receptor protein from both human and bat origin. We have also found that cell surface TLRs, especially TLR4 is most likely to be involved in recognizing molecular patterns from SARS-CoV-2 to induce inflammatory responses. The present study supported the zoonotic origin of SARS-CoV-2 from a bat and also revealed that TLR4 may have a crucial role in the virus-induced inflammatory consequences associated with COVID-19. Therefore, selective targeting of TLR4-spike protein interaction by designing competitive TLR4-antagonists could pave a new way to treat COVID-19. Finally, this study is expected to improve our understanding on the immunobiology of SARS-CoV-2 and could be useful in adopting spike protein, ACE2, or TLR-guided intervention strategy against COVID-19 shortly.


Subject(s)
Alphacoronavirus/chemistry , Angiotensin-Converting Enzyme 2/chemistry , Receptors, Virus/chemistry , SARS-CoV-2/chemistry , Spike Glycoprotein, Coronavirus/chemistry , Toll-Like Receptors/chemistry , Alphacoronavirus/classification , Alphacoronavirus/metabolism , Alphacoronavirus/pathogenicity , Angiotensin-Converting Enzyme 2/classification , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , Binding Sites , COVID-19/immunology , COVID-19/virology , Chiroptera/immunology , Chiroptera/virology , Data Mining , Eutheria/immunology , Eutheria/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Models, Molecular , Phylogeny , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, Virus/classification , Receptors, Virus/genetics , Receptors, Virus/metabolism , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/classification , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Thermodynamics , Toll-Like Receptors/classification , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Viverridae/immunology , Viverridae/virology
6.
Chest ; 155(5): 1018-1025, 2019 05.
Article in English | MEDLINE | ID: mdl-30659817

ABSTRACT

Human rhinoviruses (RVs) are picornaviruses that can cause a variety of upper and lower respiratory tract illnesses, including the common cold, bronchitis, pneumonia, and exacerbations of chronic respiratory diseases such as asthma. There are currently > 160 known types of RVs classified into three species (A, B, and C) that use three different cellular membrane glycoproteins expressed in the respiratory epithelium to enter the host cell. These viral receptors are intercellular adhesion molecule 1 (used by the majority of RV-A and all RV-B types), low-density lipoprotein receptor family members (used by 12 RV-A types), and cadherin-related family member 3 (CDHR3; used by RV-C). RV-A and RV-B interactions with intercellular adhesion molecule 1 and low-density lipoprotein receptor glycoproteins are well defined and their cellular functions have been described, whereas the mechanisms of the RV-C interaction with CDHR3 and its cellular functions are being studied. A single nucleotide polymorphism (rs6967330) in CDHR3 increases cell surface expression of this protein and, as a result, also promotes RV-C infections and illnesses. There are currently no approved vaccines or antiviral therapies available to treat or prevent RV infections, which is a major unmet medical need. Understanding interactions between RV and cellular receptors could lead to new insights into the pathogenesis of respiratory illnesses as well as lead to new approaches to control respiratory illnesses caused by RV infections.


Subject(s)
Picornaviridae Infections , Receptors, Virus , Rhinovirus , Host Microbial Interactions , Humans , Picornaviridae Infections/classification , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Receptors, Virus/classification , Receptors, Virus/physiology , Rhinovirus/classification , Rhinovirus/physiology
7.
Vopr Virusol ; 56(4): 4-8, 2011.
Article in Russian | MEDLINE | ID: mdl-21899061

ABSTRACT

The overview analyzes an update on and current concepts of the initial stage of viral infection of sensitive cells. It considers the nature of virus receptors, the mechanisms of virus-receptor interaction, methodical approaches to identifying the receptor role of cell molecules for various viruses, and the association of the initial stage of viral infection with its subsequent ones.


Subject(s)
Nucleocapsid/metabolism , Receptors, Virus , Virus Attachment , Antibody-Dependent Enhancement/physiology , Antigens, Surface/metabolism , Glycolipids/metabolism , Humans , Membrane Glycoproteins/metabolism , Receptors, Virus/classification , Receptors, Virus/genetics , Receptors, Virus/metabolism , Toll-Like Receptors/metabolism , Viral Interference/physiology , Virus Diseases/microbiology
8.
Blood ; 110(7): 2565-8, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17620455

ABSTRACT

Members of the T-cell immunoglobulin- and mucin-domain-containing molecule (TIM) family have roles in T-cell-mediated immune responses. TIM-1 and TIM-2 are predominantly expressed on T helper type 2 (Th2) cells, whereas TIM-3 is preferentially expressed on Th1 and Th17 cells. We found that TIM-1 and TIM-3, but neither TIM-2 nor TIM-4, were constitutively expressed on mouse peritoneal mast cells and bone marrow-derived cultured mast cells (BMCMCs). After IgE + Ag stimulation, TIM-1 expression was down-regulated on BMCMCs, whereas TIM-3 expression was up-regulated. We also found that recombinant mouse TIM-4 (rmTIM-4), which is a ligand for TIM-1, as well as an anti-TIM-3 polyclonal Ab, can promote interleukin-4 (IL-4), IL-6, and IL-13 production without enhancing degranulation in BMCMCs stimulated with IgE + Ag. Moreover, the anti-TIM-3 Ab, but neither anti-TIM-1 Ab nor rmTIM-4, suppressed mast-cell apoptosis. These observations suggest that TIM-1 and TIM-3 may be able to influence T-cell-mediated immune responses in part through effects on mast cells.


Subject(s)
Mast Cells/metabolism , Membrane Proteins/metabolism , Receptors, Virus/metabolism , Th2 Cells/metabolism , Animals , Cytokines/biosynthesis , Hepatitis A Virus Cellular Receptor 1 , Hepatitis A Virus Cellular Receptor 2 , Membrane Proteins/classification , Mice , Receptors, Virus/classification
9.
Nat Rev Immunol ; 6(9): 671-81, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16932752

ABSTRACT

The interaction between B- and T-lymphocyte attenuator (BTLA), an inhibitory receptor whose extracellular domain belongs to the immunoglobulin superfamily, and herpesvirus-entry mediator (HVEM), a co-stimulatory tumour-necrosis factor receptor, is unique in that it is the only receptor-ligand interaction that directly bridges these two families of receptors. This interaction has raised many questions about how receptors from two different families could interact and what downstream signalling events might occur as a result of receptor ligation. As we discuss, recent studies show that engagement of HVEM with its endogenous ligand (LIGHT) from the tumour-necrosis factor family induces a powerful immune response, whereas HVEM interactions with BTLA negatively regulate T-cell responses.


Subject(s)
Receptors, Immunologic/immunology , Receptors, Tumor Necrosis Factor/immunology , Receptors, Virus/immunology , Animals , Disease , Humans , Ligands , Protein Binding , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/chemistry , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor/classification , Receptors, Tumor Necrosis Factor, Member 14 , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/chemistry , Receptors, Virus/classification , Signal Transduction
10.
J Immunol ; 176(9): 5478-85, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16622016

ABSTRACT

CMV can cause life-threatening disease in immunodeficient hosts. Experimental infection in mice has revealed that the genetically determined natural resistance to murine CMV (MCMV) may be mediated either by direct recognition between the NK receptor Ly49H and the pathogen-encoded glycoprotein m157 or by epistatic interaction between Ly49P and the host MHC H-2D(k). Using stocks of wild-derived inbred mice as a source of genetic diversity, we found that PWK/Pas (PWK) mice were naturally resistant to MCMV. Depletion of NK cells subverted the resistance. Analysis of backcrosses to susceptible BALB/c mice revealed that the phenotype was controlled by a major dominant locus effect linked to the NK gene complex. Haplotype analysis of 41 polymorphic markers in the Ly49h region suggested that PWK mice may share a common ancestral origin with C57BL/6 mice; in the latter, MCMV resistance is dependent on Ly49H-m157 interactions. Nevertheless, PWK mice retained viral resistance against m157-defective mutant MCMV. These results demonstrate the presence of yet another NK cell-dependent viral resistance mechanism, named Cmv4, which most likely encodes for a new NK activating receptor. Identification of Cmv4 will expand our understanding of the specificity of the innate recognition of infection by NK cells.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Cytomegalovirus/physiology , Immunity, Innate/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Amino Acid Sequence , Animals , Antigens, Ly/chemistry , Antigens, Ly/genetics , Cytomegalovirus/classification , Female , Haplotypes/genetics , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Ligands , Male , Mice , Molecular Sequence Data , Multigene Family , NK Cell Lectin-Like Receptor Subfamily A , Receptors, Immunologic/metabolism , Receptors, NK Cell Lectin-Like , Receptors, Natural Killer Cell , Receptors, Virus/classification , Receptors, Virus/metabolism , Sequence Alignment
12.
Contrib Microbiol ; 10: 232-52, 2003.
Article in English | MEDLINE | ID: mdl-12530329

ABSTRACT

Large DNA viruses such as pox- and in particular herpesviruses are notorious in their ability to evade the immune system and to be maintained in the general population. Based on the accumulated knowledge reviewed in this study it is evident that important mechanisms of these actions are the acquisition and modification of host-encoded chemokines and chemokine receptors. The described viral molecules leave nothing to chance and have thoroughly and efficiently corrupted the host immune system. Through this process viruses have identified key molecules in antiviral responses by their inhibition of these or potent ways to alter an efficient antiviral response to a weak Th2-driven response. Examples here are the chemokine scavenging by US28, attractance of Th2 cells and regulatory cells by vMIP1-3 and the selective engaging of CCR8 by MC148. Important insights into viral pathology and possible targets for antiviral therapies have been provided by UL33, UL78 and in particular ORF74 and the chances are that many more will follow. In HHV8 vMIP-2 and the chemokine-binding proteins potent anti-inflammatory agents have been provided. These have already had their potential demonstrated in animal models and may in their native or modified forms represent useful therapies in humans.


Subject(s)
Chemokines/pharmacology , Poxviridae Infections/immunology , Receptors, Chemokine/metabolism , Receptors, Virus/metabolism , Viral Proteins/pharmacology , Animals , DNA Viruses/immunology , DNA Viruses/metabolism , Herpesviridae/metabolism , Herpesviridae/pathogenicity , Humans , Mammals , Receptors, Virus/classification , Receptors, Virus/genetics
13.
Curr Top Microbiol Immunol ; 269: 203-34, 2002.
Article in English | MEDLINE | ID: mdl-12224510

ABSTRACT

The ubiquitous, opportunistic pathogen human cytomegalovirus (CMV) encodes several proteins homologous to those of the host organism. Four different CMV genes encode chemokine receptor-like peptides. These genes, UL33, UL78, US27, and US28, are expressed at various stages of infection in vitro. Their functions remain largely unknown. To date, chemokine binding and signalling has only been demonstrated for the US28 gene product. Putative ligands for the other CMV-encoded chemokine receptors are discussed on basis of phylogenetic analysis. The potential roles of these receptors in virus trafficking, persistence, and immune evasion are summarized. Similarly, modulation of expression of the host chemokines IL-8, MCP-1a and RANTES in relation to viral dissemination and persistence is reviewed.


Subject(s)
Chemokines/metabolism , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Receptors, Chemokine/metabolism , Receptors, Virus/metabolism , Animals , Humans , Phylogeny , Receptors, Chemokine/classification , Receptors, Chemokine/genetics , Receptors, Virus/classification , Receptors, Virus/genetics , Virus Latency , Virus Replication
15.
Nat Struct Biol ; 7(4): 287-91, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10742172

ABSTRACT

Siderophore binding proteins play a key role in the uptake of iron in many gram-positive and gram-negative bacteria. FhuD is a soluble periplasmic binding protein that transports ferrichrome and other hydroxamate siderophores. The crystal structure of FhuD from Escherichia coli in complex with the ferrichrome homolog gallichrome has been determined at 1.9 ¿ resolution, the first structure of a periplasmic binding protein involved in the uptake of siderophores. Gallichrome is held in a shallow pocket lined with aromatic groups; Arg and Tyr side chains interact directly with the hydroxamate moieties of the siderophore. FhuD possesses a novel fold, suggesting that its mechanisms of ligand binding and release are different from other structurally characterized periplasmic ligand binding proteins.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Escherichia coli Proteins , Escherichia coli/chemistry , Ferrichrome/analogs & derivatives , Ferrichrome/metabolism , Gallic Acid/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Transport Proteins , Periplasmic Binding Proteins , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/classification , Bacterial Outer Membrane Proteins/metabolism , Binding Sites , Carrier Proteins/classification , Crystallography, X-Ray , Ligands , Membrane Proteins/classification , Models, Molecular , Molecular Sequence Data , Protein Folding , Protein Structure, Secondary , Receptors, Virus/chemistry , Receptors, Virus/classification , Receptors, Virus/metabolism , Structure-Activity Relationship
16.
J Virol ; 74(2): 619-26, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10623723

ABSTRACT

More than 10 G protein-coupled receptors (GPCRs) have been shown to act as coreceptors for infection of human immunodeficiency virus type 1 (HIV-1), HIV-2, and simian immunodeficiency virus (SIV). We have isolated HIV-1 variants infectious to primary brain-derived CD4-positive cells (BT-3 and BT-20/N) and U87/CD4 glioma cells that are resistant to T-cell line-tropic (T-tropic), macrophage-tropic (M-tropic), and T- and M-tropic (dualtropic) (X4, R5, and R5X4) HIV-1 strains. These primary brain-derived cells were also highly susceptible to HIV-2(ROD), HIV-2(SBL6669), and SIV(mndGB-1). A factor or coreceptor that determines the susceptibility of these brain-derived cells to these HIV and SIV strains has not been fully identified. To identify this coreceptor, we examined amino acid sequences of all known HIV and SIV coreceptors and noticed that tyrosine residues are well conserved in their extracellular amino-terminal domains. By this criterion, we selected 18 GPCRs as candidates of coreceptors for HIV and SIV strains infectious to these brain-derived cells. mRNA expression of an orphan GPCR, RDC1, was detected in the brain-derived cells, the C8166 T-cell line, and peripheral blood lymphocytes, all of which are susceptible to HIV-1 variants, but not in macrophages, which are resistant to them. When a CD4-expressing cell line, NP-2/CD4, which shows strict resistance to infection not only with HIV-1 but also with HIV-2 or SIV, was transduced with the RDC1 gene, the cells became highly susceptible to HIV-2 and SIV(mnd) strains but to neither M- nor T-tropic HIV-1 strains. The cells also acquired a low susceptibility to the HIV-1 variants. These findings indicate that RDC1 is a novel coreceptor for several HIV-1, HIV-2, and SIV strains which infect brain-derived cells.


Subject(s)
HIV-1/metabolism , HIV-2/metabolism , Receptors, Cell Surface/metabolism , Receptors, Chemokine , Receptors, G-Protein-Coupled , Receptors, HIV/metabolism , Receptors, Virus/metabolism , Simian Immunodeficiency Virus/metabolism , Animals , CD4 Antigens/metabolism , Cell Line , Gene Expression , HIV-1/physiology , HIV-2/physiology , Humans , Phylogeny , Receptors, CXCR , Receptors, Cell Surface/classification , Receptors, Cell Surface/genetics , Receptors, HIV/classification , Receptors, HIV/genetics , Receptors, Virus/classification , Receptors, Virus/genetics , Simian Immunodeficiency Virus/physiology , Tumor Cells, Cultured
17.
Virology ; 274(2): 278-283, 2000.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1068412

ABSTRACT

Together with the evidence that the reduced virus growth and the antiviral state induced by interferon (IFN)-ã occurring only in macrophages from resistant animals, correlated with the decrease of MHV3 binding to macrophage membrane proteins, we show here the expression of cellular and viral genes in resistant (A/J) and susceptible (BALB/c) mouse macrophages after IFN-ã activation/infection. The expression of interferon response gene 47 and interferon regulatory factor 1 genes takes place after IFN-ã activation in both macrophages, indicating their activation. The expression of the biliary glycoprotein 1a (Bgp1a, the main virus receptor) decreased only in IFN-ã-activated A/J mouse macrophages, in contrast to the expression of the Bgp2 (alternative receptor), which was not influenced by IFN-ã activation. The synthesis of both viral mRNA and virus particles was delayed only in IFN-ã-activated A/J mouse macrophages compared with susceptible BALB/c macrophages. Besides the evidence that IFN-ã may modulate the expression of the Bgp1a isoform of carcinoembryonic antigen family, these data show that IFN-ã, which induces resistance against MHV3 infection, may be involved in the down-regulation of the main viral receptor expression, a key step forward in our understanding of the molecular basis of resistance against virus infection.


Subject(s)
Animals , Mice , Infections/classification , Receptors, Virus/classification , Murine hepatitis virus , Macrophages
18.
J Virol ; 73(8): 6500-5, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10400745

ABSTRACT

Domestic cats infected with the horizontally transmitted feline leukemia virus subgroup A (FeLV-A) often produce mutants (termed FeLV-C) that bind to a distinct cell surface receptor and cause severe aplastic anemia in vivo and erythroblast destruction in bone marrow cultures. The major determinant for FeLV-C-induced anemia has been mapped to a small region of the surface envelope glycoprotein that is responsible for its receptor binding specificity. Thus, erythroblast destruction may directly or indirectly result from FeLV-C binding to its receptor. To address these issues, we functionally cloned a putative cell surface receptor for FeLV-C (FLVCR) by using a human T-lymphocyte cDNA library in a retroviral vector. Expression of the 2.0-kbp FLVCR cDNA in naturally resistant Swiss mouse fibroblasts and Chinese hamster ovary cells caused substantial susceptibility to FeLV-C but no change in susceptibilities to FeLV-B and other retroviruses. The predicted FLVCR protein contains 555 amino acids and 12 hydrophobic potential membrane-spanning sequences. Database searches indicated that FLVCR is a member of the major-facilitator superfamily of transporters and implied that it may transport an organic anion. RNA blot analyses showed that FLVCR mRNA is expressed in multiple hematopoietic lineages rather than specifically in erythroblasts. These results suggest that the targeted destruction of erythroblasts by FeLV-C may derive from their greater sensitivity to this virus rather than from a preferential susceptibility to infection.


Subject(s)
Anemia, Aplastic/virology , Carrier Proteins/classification , Leukemia Virus, Feline/metabolism , Receptors, Virus/classification , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , CHO Cells , Caenorhabditis elegans , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cats , Cricetinae , DNA, Complementary , Gene Expression , Humans , Mice , Molecular Sequence Data , Receptors, Virus/chemistry , Receptors, Virus/genetics , Sequence Homology, Amino Acid , Tissue Distribution
19.
Eur J Neurosci ; 8(2): 273-81, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8714698

ABSTRACT

A surface receptor complex of Mr approximately 65 000 (p65) and approximately 95 000 (p95) is expressed in cells of the central nervous system of mice. This receptor is recognized by monoclonal antibody 87.92.6 or by reovirus type 3 haemagglutinin as unnatural ligands. The p65/p95 receptor is expressed mostly in neural embryonic precursors undergoing proliferation, especially those in the S-G2 phase of the cell cycle. Receptor expression decreases progressively throughout embryogenesis to low but detectable levels in the adult brain. Biochemical characterization revealed that the neural p65/p95 receptor complex is indistinguishable from the p65/p95 receptor expressed in T cells, where receptor ligation leads to a mitogenic block. In neural and lymphoid tissues the p65/p95 receptor (or an associated protein) possesses a tyrosine kinase enzymatic activity. Receptor ligation in neural cells resulted in the rapid tyrosine phosphorylation of cellular proteins which are different from substrates phosphorylated in T cells. Differential substrate coupling to the receptor may account for differences in signal transduction and biology between neural cells and T cells. Further study of this receptor complex may help define important features of neural proliferation, differentiation and survival.


Subject(s)
Calcium-Binding Proteins , G2 Phase , Membrane Glycoproteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/cytology , S Phase , Animals , Antibodies, Monoclonal/immunology , Brain/cytology , Brain/embryology , Brain/growth & development , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/immunology , Neurons/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Virus/classification , Signal Transduction , Synaptotagmin I , Synaptotagmins
20.
Immunol Lett ; 48(2): 91-5, 1995 Dec.
Article in English | MEDLINE | ID: mdl-8719105

ABSTRACT

Membrane cofactor protein (MCP, CD46) of the complement system is a measles virus (MV) receptor. Human lymphocytes express a heavily glycosylated (H) and a lightly glycosylated (L) form of MCP, which confers a two-band profile on SDS-PAGE the ratio of which is controlled genetically and organ-specifically. In contrast, granulocytes express a single heavily glycosylated form regardless of lymphocyte MCP phenotype. We investigated susceptibility to MV of granulocytes and lymphocytes from individuals with different lymphocyte MCP phenotypes. In any individual, granulocytes were > 10-fold less susceptible to MV than lymphocytes, and the lymphocytes with predominant H form were generally less susceptible to those with an increasing amount of L form. Thus, lymphocytes always exhibit high susceptibility to MV compared to granulocytes in all individuals. This finding may explain the lymphopenia and immunosuppression observed secondary to MV infection.


Subject(s)
Antigens, CD/classification , Granulocytes/virology , Lymphocytes/virology , Measles virus/immunology , Membrane Glycoproteins/classification , Receptors, Virus/classification , Animals , Antigens, CD/analysis , CHO Cells , Cricetinae , Granulocytes/immunology , Humans , Immunity, Innate , Immunophenotyping , Lymphocytes/immunology , Membrane Cofactor Protein , Membrane Glycoproteins/analysis , Receptors, Virus/analysis , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL