Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 526(1): 135-140, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32199615

RESUMO

The new coronavirus (SARS-CoV-2) outbreak from December 2019 in Wuhan, Hubei, China, has been declared a global public health emergency. Angiotensin I converting enzyme 2 (ACE2), is the host receptor by SARS-CoV-2 to infect human cells. Although ACE2 is reported to be expressed in lung, liver, stomach, ileum, kidney and colon, its expressing levels are rather low, especially in the lung. SARS-CoV-2 may use co-receptors/auxiliary proteins as ACE2 partner to facilitate the virus entry. To identify the potential candidates, we explored the single cell gene expression atlas including 119 cell types of 13 human tissues and analyzed the single cell co-expression spectrum of 51 reported RNA virus receptors and 400 other membrane proteins. Consistent with other recent reports, we confirmed that ACE2 was mainly expressed in lung AT2, liver cholangiocyte, colon colonocytes, esophagus keratinocytes, ileum ECs, rectum ECs, stomach epithelial cells, and kidney proximal tubules. Intriguingly, we found that the candidate co-receptors, manifesting the most similar expression patterns with ACE2 across 13 human tissues, are all peptidases, including ANPEP, DPP4 and ENPEP. Among them, ANPEP and DPP4 are the known receptors for human CoVs, suggesting ENPEP as another potential receptor for human CoVs. We also conducted "CellPhoneDB" analysis to understand the cell crosstalk between CoV-targets and their surrounding cells across different tissues. We found that macrophages frequently communicate with the CoVs targets through chemokine and phagocytosis signaling, highlighting the importance of tissue macrophages in immune defense and immune pathogenesis.


Assuntos
Betacoronavirus/fisiologia , Receptores Virais/genética , Análise de Sequência de RNA , Análise de Célula Única , Enzima de Conversão de Angiotensina 2 , COVID-19 , Coronavirus , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Humanos , Macrófagos/metabolismo , Especificidade de Órgãos , Pandemias , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/isolamento & purificação , Peptidil Dipeptidase A/genética , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Receptores Virais/isolamento & purificação , SARS-CoV-2
2.
Protein Expr Purif ; 172: 105637, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32278001

RESUMO

With many crucial roles in enzymatic aerobic metabolism, the concentration of the heme must be tightly regulated. The heme exporter Feline Leukemia Virus sub-group C Receptor 1a (FLVCR1a), an integral membrane protein with twelve transmembrane helices, is a key player in the maintenance of cellular heme homeostasis. It was first identified as the host receptor for the Feline Leukemia Virus sub-group C (FeLV-C), a retrovirus causing hematological abnormalities in cats and other felines. Mutations in the Flvcr1 were later identified in human patients affected by Posterior Column Ataxia and Retinitis Pigmentosa (PCARP) and Hereditary Sensory and Autonomic Neuropathies (HSANs). Despite being an essential component in heme balance, currently there is a lack in the understanding of its function at the molecular level, including the effect of disease-causing mutations on protein function and structure. Therefore, there is a need for protocols to achieve efficient recombinant production yielding milligram amounts of highly pure protein to be used for biochemical and structural studies. Here, we report the first FLVCR1a reliable protocol suitable for both antibody generation and structural characterisation.


Assuntos
Proteínas de Transporte , Expressão Gênica , Heme , Proteínas de Membrana Transportadoras , Receptores Virais , Animais , Proteínas de Transporte/biossíntese , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Gatos , Humanos , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/isolamento & purificação , Camundongos , Receptores Virais/biossíntese , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/isolamento & purificação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação
3.
PLoS Pathog ; 10(10): e1004424, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25275643

RESUMO

Antibodies targeting receptor-mediated entry of HCV into hepatocytes confer limited therapeutic benefits. Evidence suggests that exosomes can transfer genetic materials between cells; however, their role in HCV infection remains obscure. Here, we show that exosomes isolated from sera of chronic HCV infected patients or supernatants of J6/JFH1-HCV-infected Huh7.5 cells contained HCV RNA. These exosomes could mediate viral receptor-independent transmission of HCV to hepatocytes. Negative sense HCV RNA, indicative of replication competent viral RNA, was present in exosomes of all HCV infected treatment non-responders and some treatment-naïve individuals. Remarkably, HCV RNA was associated with Ago2, HSP90 and miR-122 in exosomes isolated from HCV-infected individuals or HCV-infected Huh7.5 cell supernatants. Exosome-loading with a miR-122 inhibitor, or inhibition of HSP90, vacuolar H+-ATPases, and proton pumps, significantly suppressed exosome-mediated HCV transmission to naïve cells. Our findings provide mechanistic evidence for HCV transmission by blood-derived exosomes and highlight potential therapeutic strategies.


Assuntos
Exossomos/metabolismo , Hepacivirus/fisiologia , Hepatócitos/virologia , RNA Viral/genética , Proteínas Argonautas/metabolismo , Células Cultivadas , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , MicroRNAs/metabolismo , Ligação Proteica , Receptores Virais/isolamento & purificação , Replicação Viral/fisiologia
4.
Indian J Med Res ; 144(1): 38-45, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27834324

RESUMO

BACKGROUND & OBJECTIVES: It is important to understand the role of cell surface receptors in susceptibility to infectious diseases. CD155 a member of the immunoglobulin super family, serves as the poliovirus receptor (PVR). Heterozygous (Ala67Thr) polymorphism in CD155 has been suggested as a risk factor for paralytic outcome of poliovirus infection. The present study pertains to the development of a screening test to detect the single nucleotide (SNP) polymorphism in the CD155 gene. METHODS: New primers were designed for PCR, sequencing and SNP analysis of Exon2 of CD155 gene. DNAs extracted from either whole blood (n=75) or cells from oral cavity (n=75) were used for standardization and validation of the SNP assay. DNA sequencing was used as the gold standard method. RESULTS: A new SNP assay for detection of heterozygous Ala67Thr genotype was developed and validated by testing 150 DNA samples. Heterozygous CD155 was detected in 27.33 per cent (41/150) of DNA samples tested by both SNP detection assay and sequencing. INTERPRETATION & CONCLUSIONS: The SNP detection assay was successfully developed for identification of Ala67Thr polymorphism in human PVR/CD155 gene. The SNP assay will be useful for large scale screening of DNA samples.


Assuntos
Poliomielite/genética , Poliovirus/genética , Receptores Virais/genética , Análise de Sequência de DNA/métodos , Genótipo , Heterozigoto , Humanos , Poliomielite/diagnóstico , Polimorfismo de Nucleotídeo Único , Receptores Virais/isolamento & purificação
5.
Avian Pathol ; 42(1): 60-71, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23391183

RESUMO

Avian influenza (AI) viruses have been detected in more than 105 wild bird species from 12 different orders but species-related differences in susceptibility to AI viruses exist. Expression of α2,3-linked (avian-type) and α2,6-linked (human-type) sialic acid (SA) influenza virus receptors in tissues is considered one of the determinants of the host range and tissue tropism of influenza viruses. We investigated the expression of these SA receptors in 37 wild bird species from 11 different orders by lectin histochemistry. Two isoforms of Maackia amurensis (MAA) lectin, MAA1 and MAA2, were used to detect α2,3-linked SA, and Sambucus nigra lectin was used to detect α2,6-linked SA. All species evaluated expressed α2,3-linked and α2,6-linked SA receptors in endothelial cells and renal tubular epithelial cells. Both α2,3-linked and α-2,6-linked SA receptors were expressed in respiratory and intestinal tract tissues of aquatic and terrestrial wild bird species from different taxa, but differences in SA expression and in the predominant isoform of MAA lectin bound were observed. With a few possible exceptions, these observed differences were not generally predictive of reported species susceptibility to AI viruses based on published experimental and field data.


Assuntos
Vírus da Influenza A/fisiologia , Influenza Aviária/metabolismo , Lectinas/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Animais , Aves , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Especificidade de Hospedeiro , Influenza Aviária/virologia , Mucosa Intestinal/metabolismo , Intestinos/virologia , Maackia/metabolismo , Especificidade de Órgãos , Isoformas de Proteínas , Receptores de Superfície Celular/isolamento & purificação , Receptores Virais/isolamento & purificação , Sistema Respiratório/metabolismo , Sistema Respiratório/virologia , Especificidade da Espécie
6.
Proc Natl Acad Sci U S A ; 107(45): 19496-501, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974973

RESUMO

The resurrection of endogenous retroviruses from inactive molecular fossils has allowed the investigation of interactions between extinct pathogens and their hosts that occurred millions of years ago. Two such paleoviruses, chimpanzee endogenous retrovirus-1 and -2 (CERV1 and CERV2), are relatives of modern MLVs and are found in the genomes of a variety of Old World primates, but are absent from the human genome. No extant CERV1 and -2 proviruses are known to encode functional proteins. To investigate the host range restriction of these viruses, we attempted to reconstruct functional envelopes by generating consensus genes and proteins. CERV1 and -2 enveloped MLV particles infected cell lines from a range of mammalian species. Using CERV2 Env-pseudotyped MLV reporters, we identified copper transport protein 1 (CTR1) as a receptor that was presumably used by CERV2 during its ancient exogenous replication in primates. Expression of human CTR1 was sufficient to confer CERV2 permissiveness on otherwise resistant hamster cells, and CTR1 knockdown or CuCl(2) treatment specifically inhibited CERV2 infection of human cells. Mutations in highly conserved CTR1 residues that have rendered hamster cells resistant to CERV2 include a unique deletion in a copper-binding motif. These CERV2 receptor-inactivating mutations in hamster CTR1 are accompanied by apparently compensating changes, including an increased number of extracellular copper-coordinating residues, and this may represent an evolutionary barrier to the acquisition of CERV2 resistance in primates.


Assuntos
Retrovirus Endógenos/química , Extinção Biológica , Receptores Virais/isolamento & purificação , Animais , Proteínas de Transporte de Cátions/genética , Transportador de Cobre 1 , Cricetinae , Humanos , Dados de Sequência Molecular , Pan troglodytes/virologia , Vírus/genética , Vírus/patogenicidade
7.
Acta Virol ; 55(2): 93-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21692556

RESUMO

Coxsackievirus group B and adenovirus receptor (CAR) is a major receptor for the adenovirus groups that has drawn overall attention over the past decade. Although this protein could potentially be used as an agent for the blocking of adenovirus infection, large-scale production of highly purified human CAR in eukaryotic expression system has not been reported. In the present study, we showed the construction of recombinant baculovirus highly-expressing the extracellular domain of human coxsackievirus-adenovirus receptor (exCAR) in High Five insect cells. The recombinant exCAR was recovered from the cell culture medium as a secreted soluble protein and purified by Ni-NTA affinity chromatography. The final yield of recombinant exCAR was about 8-10 mg/l of supernatant with the purity of 96.3%. Binding activity assay showed that the recombinant exCAR exhibited an intact ability of binding to the knob domain of the adenovirus type 5 fiber protein (Ad fiber knob) displayed by T7 phage. These results showed that the recombinant human exCAR produced in insect cells and purified by Ni-NTA chromatography retained its ability to bind to the Ad fiber knob and could potentially be used in therapy of adenovirus infection.


Assuntos
Baculoviridae/genética , Expressão Gênica , Vetores Genéticos/genética , Receptores Virais/genética , Proteínas Recombinantes/genética , Animais , Baculoviridae/metabolismo , Linhagem Celular , Vetores Genéticos/metabolismo , Humanos , Estrutura Terciária de Proteína , Receptores Virais/química , Receptores Virais/isolamento & purificação , Receptores Virais/metabolismo , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Spodoptera
8.
Carbohydr Polym ; 260: 117797, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33712145

RESUMO

Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has resulted in a pandemic and continues to spread at an unprecedented rate around the world. Although a vaccine has recently been approved, there are currently few effective therapeutics to fight its associated disease in humans, COVID-19. SARS-CoV-2 and the related severe acute respiratory syndrome (SARS-CoV-1), and Middle East respiratory syndrome (MERS-CoV) result from zoonotic respiratory viruses that have bats as the primary host and an as yet unknown secondary host. While each of these viruses has different protein-based cell-surface receptors, each rely on the glycosaminoglycan, heparan sulfate as a co-receptor. In this study we compare, for the first time, differences and similarities in the structure of heparan sulfate in human and bat lungs. Furthermore, we show that the spike glycoprotein of COVID-19 binds 3.5 times stronger to human lung heparan sulfate than bat lung heparan sulfate.


Assuntos
Heparitina Sulfato/metabolismo , Pulmão/química , Receptores Virais/metabolismo , SARS-CoV-2/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Quirópteros , Feminino , Heparitina Sulfato/química , Heparitina Sulfato/isolamento & purificação , Humanos , Masculino , Estrutura Molecular , Peso Molecular , Ligação Proteica , Receptores Virais/química , Receptores Virais/isolamento & purificação
9.
Proc Natl Acad Sci U S A ; 104(26): 11032-7, 2007 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-17581869

RESUMO

This study demonstrates the power of a genetic selection to identify a variant virus that uses a new retroviral receptor protein. We screened a random peptide library within the receptor-binding domain of a feline leukemia virus retroviral Envelope (FeLV Env) protein for productive infection of feline AH927 cells. One variant, A5, obtained with altered tropic properties acquired the ability to use the solute carrier protein family 35 member F2 (SLC35F2) as a receptor. The SLC35F2 protein is a presumed transporter of unknown function predicted to encode 8 to 10 transmembrane-spanning regions and is not homologous to any identified retroviral receptor. Expression of the feline SLC35F2 cDNA in nonpermissive cells renders the cells susceptible to infection by A5 virus, with remarkably high titers in the range of 10(5) infectious units per ml. The human SLC35F2 ORF also functioned as the retroviral receptor, albeit at lower efficiency than the feline homologue. The successful selection of a novel molecule, the SLC35F2 transporter/channel-type protein, as a receptor by the FeLV Env backbone suggests that multipass transmembrane proteins may be particularly suited for use in productive viral entry and fusion. The analysis of retroviral Env libraries randomized in the receptor-binding domain offers a viable means to develop viral vectors targeted to specific cell types in the absence of known targeting ligands.


Assuntos
Vírus da Leucemia Felina/patogenicidade , Proteínas de Membrana Transportadoras/fisiologia , Biblioteca de Peptídeos , Receptores Virais/isolamento & purificação , Proteínas do Envelope Viral/fisiologia , Animais , Sítios de Ligação , Gatos , Linhagem Celular , Variação Genética , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Dados de Sequência Molecular , Receptores Virais/metabolismo , Retroviridae , Infecções por Retroviridae/etiologia , Infecções Tumorais por Vírus/etiologia , Proteínas do Envelope Viral/metabolismo
10.
Science ; 275(5304): 1320-3, 1997 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-9036860

RESUMO

A complementary DNA clone has been isolated that encodes a coxsackievirus and adenovirus receptor (CAR). When transfected with CAR complementary DNA, nonpermissive hamster cells became susceptible to coxsackie B virus attachment and infection. Furthermore, consistent with previous studies demonstrating that adenovirus infection depends on attachment of a viral fiber to the target cell, CAR-transfected hamster cells bound adenovirus in a fiber-dependent fashion and showed a 100-fold increase in susceptibility to virus-mediated gene transfer. Identification of CAR as a receptor for these two unrelated and structurally distinct viral pathogens is important for understanding viral pathogenesis and has implications for therapeutic gene delivery with adenovirus vectors.


Assuntos
Adenovírus Humanos/metabolismo , Enterovirus Humano B/metabolismo , Receptores Virais/isolamento & purificação , Adenovírus Humanos/genética , Adenovírus Humanos/fisiologia , Sequência de Aminoácidos , Animais , Células CHO , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Cricetinae , Efeito Citopatogênico Viral , Enterovirus Humano B/fisiologia , Técnicas de Transferência de Genes , Vetores Genéticos , Células HeLa , Humanos , Dados de Sequência Molecular , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/metabolismo , Alinhamento de Sequência , Transfecção , Replicação Viral
12.
Life Sci ; 231: 116543, 2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31176775

RESUMO

AIMS: CD155 is a ligand of the NK activating receptor DNAM-1, it has been described in a variety of human malignancies, but its expression in breast cancer remains unclear and poorly studied. MAIN METHODS: CD155 expression and NK cells infiltration were investigated in 158 patients with breast cancer by immunohistochemistry (IHC). Statistical analyses were performed to evaluate correlations of CD155 expression with clinical-pathological features, prognosis and tumor immunity. KEY FINDINGS: Tumor cytoplasmic CD155 (cyt-CD155) was associated with lymphovascular invasion (p = 0.011), and membranous CD155 (m-CD155) was strongly correlated with the presence of Tumor Infiltrating natural killer cells (NK-TILs) (p = 0.0003). Survival analysis demonstrated that patients with high cyt-CD155 had a significantly worse overall survival (p < 0.001) and death free survival (p = 0.014) than those with low expression, while high levels of m-CD155 correlated with a better prognosis (p = 0.037). Furthermore, we found that patients with m-CD155Low/NKLow tumors had a significantly reduced overall survival (p = 0.012). Multivariate analysis showed that positive tumor m-CD155 status was a significant independent marker of good prognosis. Meanwhile, high cyt-CD155 expression was identified as an independent poor prognostic predictor, suggesting a key role in this malignancy. SIGNIFICANCE: Altogether, our results revealed that cyt-CD155 was associated with invasiveness and poorer prognosis, but the concomitant presence of m-CD155 and NK-TILs had an opposite prognostic relevance in breast cancer. These results raised the importance of CD155 IHC analysis to elucidate biomarker localization, leading to better understand and design therapeutic molecule targeting CD155 in breast tumors.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Receptores Virais/genética , Receptores Virais/isolamento & purificação , Receptores Virais/metabolismo , Adulto , Idoso , Antígenos de Diferenciação de Linfócitos T/metabolismo , Neoplasias da Mama/imunologia , Citoplasma/metabolismo , Intervalo Livre de Doença , Feminino , Humanos , Imuno-Histoquímica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Prognóstico , Receptores Virais/imunologia , Estudos Retrospectivos , Análise de Sobrevida
13.
BMC Microbiol ; 8: 118, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18625079

RESUMO

BACKGROUND: Vector competence refers to the intrinsic permissiveness of an arthropod vector for infection, replication and transmission of a virus. Notwithstanding studies of Quantitative Trait Loci (QTL) that influence the ability of Aedes aegypti midgut (MG) to become infected with dengue virus (DENV), no study to date has been undertaken to identify genetic markers of vector competence. Furthermore, it is known that mosquito populations differ greatly in their susceptibility to flaviviruses. Differences in vector competence may, at least in part, be due to the presence of specific midgut epithelial receptors and their identification would be a significant step forward in understanding the interaction of the virus with the mosquito. The first interaction of DENV with the insect is through proteins in the apical membrane of the midgut epithelium resulting in binding and receptor-mediated endocytosis of the virus, and this determines cell permissiveness to infection. The susceptibility of mosquitoes to infection may therefore depend on their specific virus receptors. To study this interaction in Ae. aegypti strains that differ in their vector competence for DENV, we investigated the DS3 strain (susceptible to DENV), the IBO-11 strain (refractory to infection) and the membrane escape barrier strain, DMEB, which is infected exclusively in the midgut epithelial cells. RESULTS: (1) We determined the MG proteins that bind DENV by an overlay protein binding assay (VOPBA) in Ae. aegypti mosquitoes of the DS3, DMEB and IBO-11 strains. The main protein identified had an apparent molecular weight of 67 kDa, although the protein identified in the IBO-11 strain showed a lower mass (64 kDa). (2) The midgut proteins recognized by DENV were also determined by VOPBA after two-dimensional gel electrophoresis. (3) To determine whether the same proteins were identified in all three strains, we obtained polyclonal antibodies against R67 and R64 and tested them against the three strains by immunoblotting; both antibodies recognized the 67 and 64 kDa proteins, corroborating the VOPBA results. (4) Specific antibodies against both proteins were used for immunofluorescent location by confocal microscopy; the antibodies recognized the basal lamina all along the MG, and cell membranes and intercellular spaces from the middle to the end of the posterior midgut (pPMG) in the neighborhood of the hindgut. (5) Quantitative analysis showed more intense fluorescence in DS3 and DMEB than in IBO-11. (6) The viral envelope antigen was not homogeneously distributed during MG infection but correlated with MG density and the distribution of R67/R64. CONCLUSION: In this paper we provide evidence that the 67 kDa protein (R67/R64), described previously as a DENV receptor, is related to vector competence in Ae. aegypti. Consequently, our results strongly suggest that this protein may be a marker of vector competence for DENV in Ae. aegypti mosquitoes.


Assuntos
Aedes/genética , Aedes/virologia , Vírus da Dengue/genética , Marcadores Genéticos , Insetos Vetores/genética , Insetos Vetores/virologia , Receptores Virais/isolamento & purificação , Animais , Cromatografia de Afinidade , Eletroforese em Gel Bidimensional , Células Epiteliais/virologia , Immunoblotting , Microscopia Confocal , Peso Molecular , Receptores Virais/metabolismo , Fatores de Tempo
14.
Virol J ; 4: 83, 2007 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-17803826

RESUMO

Japanese encephalitis virus (JEV) is a member of mosquito-borne Flaviviridae. To date, the mechanisms of the early events of JEV infection remain poorly understood, and the cellular receptors are unidentified. There are evidences that the structure of the virus attachment proteins (VAP), envelope glycoprotein of mosquito-borne flaviviruses is very similar, and the vector-virus interaction of mosquito-borne flaviviruses is also very similar. Based on the studies previously demonstrated that the similar molecules present on the mosquito cells involved in the uptake process of JEV, West Nile virus (WNV) and Dengue virus (DV), it is proposed that the same receptor molecules for mosquito-borne flaviviruses (JEV, WNV and DV) may present on the surface of C6/36 mosquito cells. By co-immunoprecipitation assay, we investigated a 74-KDa protein on the C6/36 cells binds JEV, and the mass spectrometry results indicated it may be heat shock cognate protein 70(HSC70) from Aedes aegypti. Based upon some other viruses use of heat shock protein 70 (HSP70) family proteins as cell receptors, its possible HSC70's involvement in the fusion of the JEV E protein with the C6/36 cells membrane, and known form of cation channels in the interaction of HSC70 with the lipid bilayer, it will further be proposed that HSC70 as a penetration receptor mediates JEV entry into C6/36 cells.


Assuntos
Culicidae/virologia , Vírus da Dengue/fisiologia , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Proteínas de Choque Térmico HSC70/isolamento & purificação , Receptores Virais/isolamento & purificação , Ligação Viral , Vírus do Nilo Ocidental/fisiologia , Animais , Linhagem Celular , Proteínas de Choque Térmico HSC70/química , Proteínas de Choque Térmico HSC70/metabolismo , Imunoprecipitação , Espectrometria de Massas , Ligação Proteica , Receptores Virais/química , Receptores Virais/metabolismo , Internalização do Vírus
15.
DNA Seq ; 18(4): 302-6, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17541836

RESUMO

Aeromonas hydrophila is a significantly important pathogen causing major diseases in humans and fresh water fish. The outer membrane proteins (OMP) which are strong immunogens have been reported to act as adhesins aiding in the attachment of enteropathogenic bacteria. It is of interest to investigate the role of OMP in pathogenesis and their potential as vaccine candidates. In our laboratory, we cloned the gene encoding channel protein LamB porin of A. hydrophila. DNA sequence analysis revealed a full length gene of 1345 bp having a high level of homology with the lamB gene of different bacteria. Open reading frame of A. hydrophila lamB consists of a signal peptide of 25 amino acids, two protein translation start sites ATG present at the 31st and 37th base pairs, a translation termination codon, TAA at 1333rd base pair.


Assuntos
Aeromonas hydrophila/genética , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Maltose/fisiologia , Porinas/genética , Porinas/isolamento & purificação , Receptores Virais/genética , Receptores Virais/isolamento & purificação , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/química , Sequência de Bases , Clonagem Molecular , Dados de Sequência Molecular , Porinas/biossíntese , Porinas/química , Receptores Virais/biossíntese , Receptores Virais/química , Análise de Sequência de DNA , Análise de Sequência de Proteína
16.
Methods Mol Biol ; 1493: 41-56, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27787841

RESUMO

Semaphorins and their receptor plexins are large glycoproteins that are difficult to express using regular recombinant methods, and the widely used E. coli and baculovirus-insect cell systems have been inadequate for semaphorins and plexins which contain a large number of domains and are heavily modified by glycosylation. Here, we describe the expression of class 7 semaphorin (Sema7A) and the extracellular domain of its receptors PlexinC1, using the baculovirus-mediated mammalian cell gene transduction (BacMam) method. A robust mammalian cell expression gene cassette, including a highly efficient secretion signal peptide, is introduced into the baculovirus which subsequently enters mammalian cells for efficient expression in suspension cell culture. Large amount of high-infectivity BacMam viruses are needed for infecting suspended mammalian cells in large scale, to generate semaphorin and plexin proteins at an amount sufficient for binding experiments and crystallographic studies. The inclusion of serum in expression ensures the robustness of cell culture, but introduces substantial amount of contaminant proteins interfering with immobilized metal ion affinity purification, which can be overcome with a two-step purification scheme.


Assuntos
Baculoviridae/genética , Receptores de Superfície Celular/genética , Receptores Virais/metabolismo , Semaforinas/genética , Transdução Genética , Animais , Cromatografia em Gel , Células HEK293 , Humanos , Receptores de Superfície Celular/isolamento & purificação , Receptores de Superfície Celular/metabolismo , Receptores Virais/isolamento & purificação , Semaforinas/isolamento & purificação , Células Sf9
17.
mBio ; 8(1)2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28196955

RESUMO

The Hom-1 vesivirus was reported in 1998 following the inadvertent transmission of the animal calicivirus San Miguel sea lion virus to a human host in a laboratory. We characterized the Hom-1 strain and investigated the mechanism by which human cells could be infected. An expression library of 3,559 human plasma membrane proteins was screened for reactivity with Hom-1 virus-like particles, and a single interacting protein, human junctional adhesion molecule 1 (hJAM1), was identified. Transient expression of hJAM1 conferred susceptibility to Hom-1 infection on nonpermissive Chinese hamster ovary (CHO) cells. Virus infection was markedly inhibited when CHO cells stably expressing hJAM were pretreated with anti-hJAM1 monoclonal antibodies. Cell lines of human origin were tested for growth of Hom-1, and efficient replication was observed in HepG2, HuH7, and SK-CO15 cells. The three cell lines (of hepatic or intestinal origin) were confirmed to express hJAM1 on their surface, and clustered regularly interspaced short palindromic repeats/Cas9-mediated knockout of the hJAM1 gene in each line abolished Hom-1 propagation. Taken together, our data indicate that entry of the Hom-1 vesivirus into these permissive human cell lines is mediated by the plasma membrane protein hJAM1 as a functional receptor.IMPORTANCE Vesiviruses, such as San Miguel sea lion virus and feline calicivirus, are typically associated with infection in animal hosts. Following the accidental infection of a laboratory worker with San Miguel sea lion virus, a related virus was isolated in cell culture and named Hom-1. In this study, we found that Hom-1 could be propagated in a number of human cell lines, making it the first calicivirus to replicate efficiently in cultured human cells. Screening of a library of human cell surface membrane proteins showed that the virus could utilize human junctional adhesion molecule 1 as a receptor to enter cells and initiate replication. The Hom-1 virus presents a new system for the study of calicivirus biology and species specificity.


Assuntos
Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Vesivirus/fisiologia , Replicação Viral , Animais , Células CHO , Gatos , Moléculas de Adesão Celular/deficiência , Moléculas de Adesão Celular/isolamento & purificação , Membrana Celular/química , Membrana Celular/genética , Cricetinae , Cricetulus , Humanos , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/isolamento & purificação , Receptores Virais/isolamento & purificação , Vesivirus/crescimento & desenvolvimento
18.
Structure ; 8(11): 1147-55, 2000 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-11080637

RESUMO

BACKGROUND: The coxsackievirus and adenovirus receptor (CAR) comprises two extracellular immunoglobulin domains, a transmembrane helix and a C-terminal intracellular domain. The amino-terminal immunoglobulin domain (D1) of CAR is necessary and sufficient for adenovirus binding, whereas the site of coxsackievirus attachment has not yet been localized. The normal cellular role of CAR is currently unknown, although CAR was recently proposed to function as a homophilic cell adhesion molecule. RESULTS: The human CAR D1 domain was bacterially expressed and crystallized. The structure was solved by molecular replacement using the structure of CAR D1 bound to the adenovirus type 12 fiber head and refined to 1.7 A resolution, including individual anisotropic temperature factors. The two CAR D1 structures are virtually identical, apart from the BC, C"D, and FG loops that are involved both in fiber head binding and homodimerization in the crystal. Analytical equilibrium ultracentrifugation shows that a dimer also exists in solution, with a dissociation constant of 16 microM. CONCLUSIONS: The CAR D1 domain forms homodimers in the crystal using the same GFCC'C" surface that interacts with the adenovirus fiber head. The homodimer is very similar to the CD2 D1-CD58 D1 heterodimer. CAR D1 also forms dimers in solution with a dissociation constant typical of other cell adhesion complexes. These results are consistent with reports that CAR may function physiologically as a homophilic cell adhesion molecule in the developing mouse brain. Adenovirus may thus have recruited an existing and conserved interaction surface of CAR to use for its own cell attachment.


Assuntos
Receptores Virais/química , Adenovírus Humanos/metabolismo , Sequência de Aminoácidos , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Cristalografia por Raios X , Dimerização , Enterovirus/metabolismo , Humanos , Ligação de Hidrogênio , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína , Receptores Virais/isolamento & purificação , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Solubilidade , Relação Estrutura-Atividade , Ultracentrifugação
19.
Mol Immunol ; 42(4): 463-9, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15607800

RESUMO

The capability of NK lymphocytes to kill tumor cells depends on different receptors/ligands interactions. In order to identify the cellular ligands recognized by "orphan" triggering receptors, mice were immunized with NK susceptible target cells. mAbs were selected that inhibited NK cytotoxicity and recognized two different molecules of 70 and 60-65 kDa. Tryptic digestion and mass spectra analysis of purified proteins identified these molecules as PVR and Nectin-2, respectively. PVR-Fc and Nectin-2-Fc chimeric molecules stained COS-7 cells expressing the DNAM-1 activating receptor and conversely, PVR and Nectin-2 CHO-K cell transfectants were stained by DNAM-1-Fc. Thus, both PVR and Nectin-2 represent specific ligands for DNAM-1. Importantly, the specific interaction between DNAM-1 (in NK cells) and PVR or Nectin-2 (in target cells) enhanced the NK-mediated lysis of tumor cells that was downregulated by mAb-mediated masking of the receptor or its ligands.


Assuntos
Antígenos de Diferenciação de Linfócitos T/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Matadoras Naturais/imunologia , Proteínas de Membrana/metabolismo , Neoplasias/imunologia , Receptores Virais/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Células COS , Moléculas de Adesão Celular/isolamento & purificação , Chlorocebus aethiops , Citotoxicidade Imunológica , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Ligantes , Proteínas de Membrana/isolamento & purificação , Camundongos , Nectinas , Mapeamento de Peptídeos , Receptores Virais/isolamento & purificação , Proteínas Recombinantes de Fusão/imunologia
20.
Biochim Biophys Acta ; 1067(1): 89-96, 1991 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-1868106

RESUMO

Wild-type and 16 variant maltoporins with site-directed cysteine substitutions at 14 sites were purified by a novel one-step affinity-chromatographic procedure. The trimer stability of purified proteins with C22S, C38S and G103C substitutions was reduced compared to wild-type maltoporin. Quantitative labelling with N-ethyl[14C]maleimide, cross-linking with bifunctional bismaleimides and disulphide formation was used to test the reactivity of cysteines in the folded protein. The maleimide reactivity of the residues was in the order: 152 approximately equal to 153 greater than 265 greater than 30 approximately equal to 103 approximately equal to 120 approximately equal to 154 approximately equal to 382 greater than 57 approximately equal to 146, with the other sites (22, 38, 97, 184) poorly labelled. Only cysteines at 152 or 153 permitted the formation of inter-subunit disulphide bonds suggesting these residues are located within 0.5-0.9 nm of each other in homotrimers of maltoporin. S152C and S153C as well as S154C permitted the formation of inter-subunit cross-links using bifunctional bismaleimides. The cross-linkability and the high reactivity to N-ethylmaleimide of the 150 region was consistent with the current model of the structure of maltoporin in the outer membrane; the reactivity of the other sites is also discussed within the context of this model.


Assuntos
Proteínas da Membrana Bacteriana Externa/análise , Cisteína , Escherichia coli/análise , Receptores Virais/isolamento & purificação , Compostos de Sulfidrila/análise , Sequência de Aminoácidos , Cromatografia de Afinidade , Etilmaleimida/farmacologia , Dados de Sequência Molecular , Mutagênese , Oxirredução , Porinas , Receptores Virais/genética , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA