Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(2): e1012022, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38359079

RESUMO

Enterovirus A71 (EV-A71) infection involves a variety of receptors. Among them, two transmembrane protein receptors have been investigated in detail and shown to be critical for infection: P-selectin glycoprotein ligand-1 (PSGL-1) in lymphocytes (Jurkat cells), and scavenger receptor class B member 2 (SCARB2) in rhabdomyosarcoma (RD) cells. PSGL-1 and SCARB2 have been reported to be expressed on the surface of Jurkat and RD cells, respectively. In the work reported here, we investigated the roles of PSGL-1 and SCARB2 in the process of EV-A71 entry. We first examined the expression of SCARB2 in Jurkat cells, and detected it within the cytoplasm, but not on the cell surface. Further, using PSGL-1 and SCARB2 knockout cells, we found that although both PSGL-1 and SCARB2 are essential for virus infection of Jurkat cells, virus attachment to these cells requires only PSGL-1. These results led us to evaluate the cell surface expression and the roles of SCARB2 in other EV-A71-susceptible cell lines. Surprisingly, in contrast to the results of previous studies, we found that SCARB2 is absent from the surface of RD cells and other susceptible cell lines we examined, and that although SCARB2 is essential for infection of these cells, it is dispensable for virus attachment. These results indicate that a receptor other than SCARB2 is responsible for virus attachment to the cell and probably for internalization of virions, not only in Jurkat cells but also in RD cells and other EV-A71-susceptible cells. SCARB2 is highly concentrated in lysosomes and late endosomes, where it is likely to trigger acid-dependent uncoating of virions, the critical final step of the entry process. Our results suggest that the essential interactions between EV-A71 and SCARB2 occur, not at the cell surface, but within the cell.


Assuntos
Enterovirus Humano A , Infecções por Enterovirus , Enterovirus , Humanos , Enterovirus/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Membrana Celular/metabolismo , Linhagem Celular , Receptores Depuradores/genética , Receptores Depuradores/metabolismo , Glicoproteínas de Membrana Associadas ao Lisossomo/genética
2.
J Neurosci ; 36(37): 9558-71, 2016 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-27629708

RESUMO

UNLABELLED: Although we are beginning to understand the late stage of neurodegenerative diseases, the molecular defects associated with the initiation of impaired cognition are poorly characterized. Here, we demonstrate that in the adult brain, the coxsackievirus and adenovirus receptor (CAR) is located on neuron projections, at the presynapse in mature neurons, and on the soma of immature neurons in the hippocampus. In a proinflammatory or diseased environment, CAR is lost from immature neurons in the hippocampus. Strikingly, in hippocampi of patients at early stages of late-onset Alzheimer's disease (AD), CAR levels are significantly reduced. Similarly, in triple-transgenic AD mice, CAR levels in hippocampi are low and further reduced after systemic inflammation. Genetic deletion of CAR from the mouse brain triggers deficits in adult neurogenesis and synapse homeostasis that lead to impaired hippocampal plasticity and cognitive deficits. We propose that post-translational CAR loss of function contributes to cognitive defects in healthy and diseased-primed brains. SIGNIFICANCE STATEMENT: This study addressed the role of the coxsackievirus and adenovirus receptor (CAR), a single-pass cell adhesion molecule, in the adult brain. Our results demonstrate that CAR is expressed by mature neurons throughout the brain. In addition, we propose divergent roles for CAR in immature neurons, during neurogenesis, and at the mature synapse. Notably, CAR loss of function also affects hippocampal plasticity.


Assuntos
Doença de Alzheimer/patologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/deficiência , Hipocampo/patologia , Neurogênese/genética , Plasticidade Neuronal/genética , Sinapses/metabolismo , Fatores Etários , Doença de Alzheimer/complicações , Doença de Alzheimer/genética , Animais , Células Cultivadas , Transtornos Cognitivos/etiologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Regulação da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Nestina/genética , Nestina/metabolismo
3.
J Virol ; 90(12): 5601-5610, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27030267

RESUMO

UNLABELLED: The coxsackievirus and adenovirus receptor (CAR) is a member of the immunoglobulin superfamily (IgSF) and functions as a receptor for coxsackie B viruses (CVBs). The extracellular portion of CAR comprises two glycosylated immunoglobulin-like domains, D1 and D2. CAR-D1 binds to the virus and is essential for virus infection; however, it is not known whether D2 is also important for infection, and the role of glycosylation has not been explored. To understand the function of these structural components in CAR-mediated CVB3 infection, we generated a panel of human (h) CAR deletion and substitution mutants and analyzed their functionality as CVB receptors, examining both virus binding and replication. Lack of glycosylation of the CAR-D1 or -D2 domains did not adversely affect CVB3 binding or infection, indicating that the glycosylation of CAR is not required for its receptor functions. Deletion of the D2 domain reduced CVB3 binding, with a proportionate reduction in the efficiency of virus infection. Replacement of D2 with the homologous D2 domain from chicken CAR, or with the heterologous type C2 immunoglobulin-like domain from IgSF11, another IgSF member, fully restored receptor function; however, replacement of CAR-D2 with domains from CD155 or CD80 restored function only in part. These data indicate that glycosylation of the extracellular domain of hCAR plays no role in CVB3 receptor function and that CAR-D2 is not specifically required. The D2 domain may function largely as a spacer permitting virus access to D1; however, the data may also suggest that D2 affects virus binding by influencing the conformation of D1. IMPORTANCE: An important step in virus infection is the initial interaction of the virus with its cellular receptor. Although the role in infection of the extracellular CAR-D1, cytoplasmic, and transmembrane domains have been analyzed extensively, nothing is known about the function of CAR-D2 and the extracellular glycosylation of CAR. Our data indicate that glycosylation of the extracellular CAR domain has only minor importance for the function of CAR as CVB3 receptor and that the D2 domain is not essential per se but contributes to receptor function by promoting the exposure of the D1 domain on the cell surface. These results contribute to our understanding of the coxsackievirus-receptor interactions.


Assuntos
Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/química , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/metabolismo , Enterovirus Humano B/fisiologia , Ligação Viral , Animais , Células CHO , Galinhas , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/genética , Cricetulus , Enterovirus Humano B/química , Glicosilação , Células HeLa , Humanos , Domínios de Imunoglobulina/genética , Mutação , Replicação Viral
4.
PLoS Pathog ; 11(10): e1005184, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26430888

RESUMO

NF449, a sulfated compound derived from the antiparasitic drug suramin, was previously reported to inhibit infection by enterovirus A71 (EV-A71). In the current work, we found that NF449 inhibits virus attachment to target cells, and specifically blocks virus interaction with two identified receptors--the P-selectin ligand, PSGL-1, and heparan sulfate glycosaminoglycan--with no effect on virus binding to a third receptor, the scavenger receptor SCARB2. We also examined a number of commercially available suramin analogues, and newly synthesized derivatives of NF449; among these, NF110 and NM16, like NF449, inhibited virus attachment at submicromolar concentrations. PSGL-1 and heparan sulfate, but not SCARB2, are both sulfated molecules, and their interaction with EV-A71 is thought to involve positively charged capsid residues, including a conserved lysine at VP1-244, near the icosahedral 5-fold vertex. We found that mutation of VP1-244 resulted in resistance to NF449, suggesting that this residue is involved in NF449 interaction with the virus capsid. Consistent with this idea, NF449 and NF110 prevented virus interaction with monoclonal antibody MA28-7, which specifically recognizes an epitope overlapping VP1-244 at the 5-fold vertex. Based on these observations we propose that NF449 and related compounds compete with sulfated receptor molecules for a binding site at the 5-fold vertex of the EV-A71 capsid.


Assuntos
Antivirais/farmacologia , Benzenossulfonatos/farmacologia , Infecções por Enterovirus/virologia , Heparitina Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Ligação Viral/efeitos dos fármacos , Sítios de Ligação , Capsídeo/química , Capsídeo/efeitos dos fármacos , Capsídeo/metabolismo , Enterovirus Humano A/efeitos dos fármacos , Enterovirus Humano A/metabolismo , Infecções por Enterovirus/metabolismo , Células HeLa , Humanos , Células Jurkat , Modelos Moleculares , Dados de Sequência Molecular , Suramina/análogos & derivados
5.
Cell Host Microbe ; 18(2): 221-32, 2015 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-26269957

RESUMO

Receptor interacting protein kinase-3 (RIP3) is an essential kinase for necroptotic cell death signaling and has been implicated in antiviral cell death signaling upon DNA virus infection. Here, we performed high-throughput RNAi screening and identified RIP3 as a positive regulator of coxsackievirus B3 (CVB) replication in intestinal epithelial cells (IECs). RIP3 regulates autophagy, a process utilized by CVB for viral replication factory assembly, and depletion of RIP3 inhibits autophagic flux and leads to the accumulation of autophagosomes and amphisomes. Additionally, later in infection, RIP3 is cleaved by the CVB-encoded cysteine protease 3C(pro), which serves to abrogate RIP3-mediated necrotic signaling and induce a nonnecrotic form of cell death. Taken together, our results show that temporal targeting of RIP3 allows CVB to benefit from its roles in regulating autophagy while inhibiting the induction of necroptotic cell death.


Assuntos
Autofagia , Cisteína Endopeptidases/metabolismo , Enterovirus Humano B/fisiologia , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Proteases Virais 3C , Células CACO-2 , Enterovirus Humano B/metabolismo , Inativação Gênica , Testes Genéticos , Humanos , Interferência de RNA
6.
J Virol ; 89(8): 4311-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25653430

RESUMO

UNLABELLED: In vitro, infection of polarized human intestinal epithelial cells by coxsackievirus B3 (CVB3) depends on virus interaction with decay-accelerating factor (DAF), a receptor expressed on the apical cell surface. Although mice are highly susceptible to CVB3 infection when virus is delivered by intraperitoneal injection, infection by the enteral route is very inefficient. Murine DAF, unlike human DAF, does not bind virus, and we hypothesized that the absence of an accessible receptor on the intestinal surface is an important barrier to infection by the oral route. We generated transgenic mice that express human DAF specifically on intestinal epithelium and measured their susceptibility to infection by a DAF-binding CVB3 isolate. Human DAF permitted CVB3 to bind to the intestinal surface ex vivo and to infect polarized monolayers of small-intestinal epithelial cells derived from DAF transgenic mice. However, expression of human DAF did not facilitate infection by the enteral route either in immunocompetent animals or in animals deficient in the interferon alpha/beta receptor. These results indicate that the absence of an apical receptor on intestinal epithelium is not the major barrier to infection of mice by the oral route. IMPORTANCE: CVB3 infection of human intestinal epithelial cells depends on DAF at the apical cell surface, and expression of human DAF on murine intestinal epithelial cells permits their infection in vitro. However, expression of human DAF on the intestinal surface of transgenic mice did not facilitate infection by the oral route. Although the role of intestinal DAF in human infection has not been directly examined, these results suggest that DAF is not the critical factor in mice.


Assuntos
Antígenos CD55/metabolismo , Suscetibilidade a Doenças/virologia , Enterovirus Humano B/fisiologia , Infecções por Enterovirus/metabolismo , Mucosa Intestinal/metabolismo , Animais , Primers do DNA/genética , Imunofluorescência , Humanos , Immunoblotting , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Virol ; 89(2): 1324-8, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25392210

RESUMO

UNLABELLED: Many coxsackievirus B (CVB) isolates bind to human decay-accelerating factor (DAF) as well as to the coxsackievirus and adenovirus receptor (CAR). However, the virus does not interact with murine DAF. To understand why CVB3 binds specifically to human DAF, we constructed a series of chimeric molecules in which specific regions of the human DAF molecule were replaced by the corresponding murine sequences. We found that replacement of human short consensus repeat 2 (SCR2) with murine SCR2 ablated virus binding to human DAF, as did deletion of human SCR2. Although replacement of human SCR4 had a partial inhibitory effect, deletion of SCR4 had no effect. Within human SCR2, replacement of serine 104 (S104) with the proline residue found in murine DAF eliminated virus binding. On the basis of the structure of the CVB3-DAF complex determined by cryo-electron microscopy, DAF S104 is in close contact with a viral capsid residue, a threonine at VP1 position 271. Replacement of this capsid residue with larger amino acids specifically eliminated virus attachment to human DAF but had no effect on attachment to CAR or replication in HeLa cells. Taken together, these results support the current model of virus-DAF interaction and point to a specific role for VP1 T271 and DAF S104 at the virus-DAF interface. IMPORTANCE: The results of the present study point to a specific role for VP1 T271 and DAF S104 at the interface between CVB3 and DAF, and they demonstrate how subtle structural changes can dramatically influence virus-receptor interactions. In addition, the results support a recent pseudoatomic model of the CVB3-DAF interaction obtained by cryo-electron microscopy.


Assuntos
Antígenos CD55/metabolismo , Proteínas do Capsídeo/metabolismo , Enterovirus Humano B/fisiologia , Interações Hospedeiro-Patógeno , Receptores Virais/metabolismo , Ligação Viral , Sequência de Aminoácidos , Animais , Antígenos CD55/genética , Linhagem Celular , Análise Mutacional de DNA , Humanos , Camundongos , Dados de Sequência Molecular , Receptores Virais/genética
8.
J Virol ; 88(1): 434-43, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24155402

RESUMO

Echovirus 7 enters polarized Caco-2 intestinal epithelial cells by a clathrin-mediated endocytic process and then moves through the endosomal system before releasing its genome into the cytoplasm. We examined the possible role in virus entry of core components of the autophagy machinery. We found that depletion of Beclin-1, Atg12, Atg14, Atg16, or LC3 with specific small interfering RNAs inhibited echovirus 7 infection upstream of uncoating but had little or no effect on virus attachment to the cell surface. These data indicate that multiple autophagy-related proteins are important for one or more events that occur after the virus has bound its receptor on the cell surface but before RNA is released from the virus capsid. Although we have not determined the mechanism by which each protein contributes to virus entry, we found that stable depletion of Atg16L1 interfered with virus internalization from the cell surface rather than with intracellular trafficking. Autophagy gene products may thus participate in the endocytic process that moves virus into polarized Caco-2 cells.


Assuntos
Autofagia , Enterovirus Humano B/fisiologia , Mucosa Intestinal/virologia , Fusão de Membrana/fisiologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Relacionadas à Autofagia , Proteína Beclina-1 , Células CACO-2 , Endossomos/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo
9.
PLoS Pathog ; 9(7): e1003511, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935488

RESUMO

Some strains of enterovirus 71 (EV71), but not others, infect leukocytes by binding to a specific receptor molecule: the P-selectin glycoprotein ligand-1 (PSGL-1). We find that a single amino acid residue within the capsid protein VP1 determines whether EV71 binds to PSGL-1. Examination of capsid sequences of representative EV71 strains revealed that the PSGL-1-binding viruses had either a G or a Q at residue 145 within the capsid protein VP1 (VP1-145G or Q), whereas PSGL-1-nonbinding viruses had VP1-145E. Using site-directed mutagenesis we found that PSGL-1-binding strains lost their capacity to bind when VP1-145G/Q was replaced by E; conversely, nonbinding strains gained the capacity to bind PSGL-1 when VP1-145E was replaced with either G or Q. Viruses with G/Q at VP1-145 productively infected a leukocyte cell line, Jurkat T-cells, whereas viruses with E at this position did not. We previously reported that EV71 binds to the N-terminal region of PSGL-1, and that binding depends on sulfated tyrosine residues within this region. We speculated that binding depends on interaction between negatively charged sulfate groups and positively charged basic residues in the virus capsid. VP1-145 on the virus surface is in close proximity to conserved lysine residues at VP1-242 and VP1-244. Comparison of recently published crystal structures of EV71 isolates with either Q or E at VP1-145 revealed that VP1-145 controls the orientation of the lysine side-chain of VP1-244: with VP1-145Q the lysine side chain faces outward, but with VP1-145E, the lysine side chain is turned toward the virus surface. Mutation of VP1-244 abolished virus binding to PSGL-1, and mutation of VP1-242 greatly reduced binding. We propose that conserved lysine residues on the virus surface are responsible for interaction with sulfated tyrosine residues at the PSGL-1 N-terminus, and that VP1-145 acts as a switch, controlling PSGL-1 binding by modulating the exposure of VP1-244K.


Assuntos
Proteínas do Capsídeo/metabolismo , Enterovirus Humano A/imunologia , Leucócitos/imunologia , Glicoproteínas de Membrana/metabolismo , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sítios de Ligação , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Sequência Conservada , Enterovirus Humano A/fisiologia , Interações Hospedeiro-Patógeno , Humanos , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo , Células Jurkat , Leucócitos/metabolismo , Leucócitos/virologia , Lisina/química , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Tirosina/análogos & derivados , Tirosina/química , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética , Ligação Viral
10.
Adv Exp Med Biol ; 790: 24-41, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23884584

RESUMO

The essential event in picornavirus entry is the delivery of the RNA genome to the cytoplasm of a target cell, where replication occurs. In the past several years progress has been made in understanding the structural changes in the virion important for uncoating and RNA release. In addition, for several viruses the endocytic mechanisms responsible for internalization have been identified, as have the cellular sites at which uncoating occurs. It has become clear that entry is not a passive process, and that viruses initiate specific signals required for entry. And we have begun to recognize that for a given virus, there may be multiple routes of entry, depending on the particular target cell and the receptors available on that cell.


Assuntos
Picornaviridae/fisiologia , Internalização do Vírus , Animais , Antígenos CD55/fisiologia , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus/fisiologia , Endocitose , Humanos , Picornaviridae/ultraestrutura , Receptores Virais/fisiologia , Ligação Viral
11.
J Virol ; 87(16): 8884-95, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23740983

RESUMO

Enteroviruses invade their hosts by crossing the intestinal epithelium. We have examined the mechanism by which echovirus 1 (EV1) enters polarized intestinal epithelial cells (Caco-2). Virus binds to VLA-2 on the apical cell surface and moves rapidly to early endosomes. Using inhibitory drugs, dominant negative mutants, and small interfering RNAs (siRNAs) to block specific endocytic pathways, we found that virus entry requires dynamin GTPase and membrane cholesterol but is independent of both clathrin- and caveolin-mediated endocytosis. Instead, infection requires factors commonly associated with macropinocytosis, including amiloride-sensitive Na(+)/H(+) exchange, protein kinase C, and C-terminal-binding protein-1 (CtBP1); furthermore, EV1 accumulates rapidly in intracellular vesicles with dextran, a fluid-phase marker. These results suggest a role for macropinocytosis in the process by which EV1 enters polarized cells to initiate infection.


Assuntos
Colesterol/metabolismo , Dinaminas/metabolismo , Enterovirus Humano B/fisiologia , Células Epiteliais/fisiologia , Interações Hospedeiro-Patógeno , Pinocitose , Internalização do Vírus , Oxirredutases do Álcool/metabolismo , Células CACO-2 , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/virologia , Humanos , Proteína Quinase C/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Vesículas Transportadoras/virologia
12.
J Virol ; 86(23): 12571-81, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22973031

RESUMO

The coxsackievirus-adenovirus receptor (CAR) and decay-accelerating factor (DAF) have been identified as cellular receptors for coxsackievirus B3 (CVB3). The first described DAF-binding isolate was obtained during passage of the prototype strain, Nancy, on rhabdomyosarcoma (RD) cells, which express DAF but very little CAR. Here, the structure of the resulting variant, CVB3-RD, has been solved by X-ray crystallography to 2.74 Å, and a cryo-electron microscopy reconstruction of CVB3-RD complexed with DAF has been refined to 9.0 Å. This new high-resolution structure permits us to correct an error in our previous view of DAF-virus interactions, providing a new footprint of DAF that bridges two adjacent protomers. The contact sites between the virus and DAF clearly encompass CVB3-RD residues recently shown to be required for binding to DAF; these residues interact with DAF short consensus repeat 2 (SCR2), which is known to be essential for virus binding. Based on the new structure, the mode of the DAF interaction with CVB3 differs significantly from the mode reported previously for DAF binding to echoviruses.


Assuntos
Antígenos CD55/química , Enterovirus Humano B/ultraestrutura , Modelos Moleculares , Conformação Proteica , Receptores Virais/química , Antígenos CD55/metabolismo , Linhagem Celular Tumoral , Microscopia Crioeletrônica , Cristalização , Cristalografia por Raios X , Humanos , Receptores Virais/metabolismo
13.
mBio ; 3(2)2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496312

RESUMO

UNLABELLED: Enteroviruses invade the host by crossing the intestinal mucosa, which is lined by polarized epithelium. A number of enteroviruses, including echoviruses (EV) and group B coxsackieviruses (CVB), initiate infection by attaching to decay-accelerating factor (DAF), a molecule that is highly expressed on the apical surface of polarized epithelial cells. We previously observed that entry of DAF-binding CVB3 into polarized intestinal epithelial cells occurs by an unusual endocytic mechanism that requires caveolin but does not involve clathrin or dynamin. Here we examined the entry of a DAF-binding echovirus, EV7. We found that drugs, small interfering RNAs (siRNAs), and dominant negative mutants that target factors required for clathrin-mediated endocytosis, including clathrin and dynamin, inhibited both EV7 infection and internalization of virions from the cell surface. Once virus had entered the cell, it colocalized with markers of early endosomes (EEA1) and then late endosomes (LAMP-2). Inhibition of endosomal maturation-with siRNAs or dominant negative mutants targeting Rab5 and Rab7-inhibited infection and prevented release of viral RNA into the cell. These results indicate that EV7 is internalized by clathrin-mediated endocytosis and then moves to early and late endosomes before releasing its RNA. Trafficking through endosomes is known to be important for viruses that depend on low pH or endosomal cathepsin proteases to complete the entry process. However, we found that EV7 infection required neither low pH nor cathepsins. IMPORTANCE: The results demonstrate that echovirus 7 (EV7), after binding to decay-accelerating factor (DAF) on the cell surface, enters cells by clathrin-mediated endocytosis; this entry mechanism differs markedly from that of another DAF-binding enterovirus, coxsackievirus B3 (CVB3). Thus, after attachment to the same cell surface receptor, these closely related viruses enter the same cells by different mechanisms. The cellular cues required for release of viral RNA from the enterovirus capsid ("uncoating") remain poorly defined. We found that EV7 moved to late endosomes and that release of RNA depended on endosomal maturation; nonetheless, EV7 did not depend on the endosomal factors implicated in uncoating and entry by other viruses. The results suggest either that an unidentified endosomal factor is essential for uncoating of EV7 or that trafficking through the endosome is an essential step in a pathway that leads to another intracellular organelle where uncoating is completed.


Assuntos
Clatrina/metabolismo , Endocitose , Enterovirus/patogenicidade , Células Epiteliais/fisiologia , Células Epiteliais/virologia , Internalização do Vírus , Proteínas rab de Ligação ao GTP/metabolismo , Antígenos CD55/metabolismo , Células CACO-2 , Endossomos/virologia , Humanos , Ligação Proteica , Proteínas rab5 de Ligação ao GTP/metabolismo , proteínas de unión al GTP Rab7
14.
Mol Biol Cell ; 22(17): 3010-21, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21737691

RESUMO

Coxsackievirus B (CVB), a member of the enterovirus family, targets the polarized epithelial cells lining the intestinal tract early in infection. Although the polarized epithelium functions as a protective barrier, this barrier is likely exploited by CVB to promote viral entry and subsequent egress. Here we show that, in contrast to nonpolarized cells, CVB-infected polarized intestinal Caco-2 cells undergo nonapoptotic necrotic cell death triggered by inositol 1,4,5-trisphosphate receptor-dependent calcium release. We further show that CVB-induced cellular necrosis depends on the Ca(2+)-activated protease calpain-2 and that this protease is involved in CVB-induced disruption of the junctional complex and rearrangements of the actin cytoskeleton. Our study illustrates the cell signaling pathways hijacked by CVB, and perhaps other viral pathogens, to promote their replication and spread in polarized cell types.


Assuntos
Sinalização do Cálcio , Calpaína/metabolismo , Enterovirus Humano B/fisiologia , Infecções por Enterovirus/virologia , Mucosa Intestinal/virologia , Necrose/virologia , Liberação de Vírus , Células CACO-2 , Proteínas do Capsídeo/metabolismo , Caspase 3/metabolismo , Polaridade Celular , Citoesqueleto/metabolismo , Retículo Endoplasmático/metabolismo , Ensaios Enzimáticos , Células HeLa , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mucosa Intestinal/enzimologia , Mucosa Intestinal/patologia , Microscopia de Fluorescência , Necrose/metabolismo , Junções Íntimas/metabolismo , Fosfolipases Tipo C/metabolismo
15.
J Virol ; 85(14): 7436-43, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21561916

RESUMO

Many coxsackievirus B isolates bind to human decay-accelerating factor (DAF) as well as to the coxsackievirus and adenovirus receptor (CAR). The first-described DAF-binding isolate, coxsackievirus B3 (CB3)-RD, was obtained during passage of the prototype strain CB3-Nancy on RD cells, which express DAF but very little CAR. CB3-RD binds to human DAF, whereas CB3-Nancy does not. To determine the molecular basis for the specific interaction of CB3-RD with DAF, we produced cDNA clones encoding both CB3-RD and CB3-Nancy and mutated each of the sites at which the RD and Nancy sequences diverged. We found that a single amino acid change, the replacement of a glutamate within VP3 (VP3-234E) with a glutamine residue (Q), conferred upon CB3-Nancy the capacity to bind DAF and to infect RD cells. Readaptation of molecularly cloned CB3-Nancy to RD cells selected for a new virus with the same VP3-234Q residue. In experiments with CB3-H3, another virus isolate that does not bind measurably to DAF, adaptation to RD cells resulted in a DAF-binding isolate with a single amino acid change within VP2 (VP2-138 N to D). Both VP3-234Q and VP2-138D were required for binding of CB3-RD to DAF. In the structure of the CB3-RD-DAF complex determined by cryo-electron microscopy, both VP3-234Q and VP2-138D are located at the contact site between the virus and DAF.


Assuntos
Substituição de Aminoácidos , Capsídeo , Enterovirus Humano B/fisiologia , Sequência de Aminoácidos , Sequência de Bases , Primers do DNA , Enterovirus Humano B/química , Imunofluorescência , Células HeLa , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
16.
J Virol ; 83(21): 11064-77, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19710132

RESUMO

Group B coxsackieviruses (CVB) use the CVB and adenovirus receptor (CAR) to enter and infect cells. Some CVB also bind to decay-accelerating factor (DAF), but that interaction alone is insufficient for infection. We previously found that CVB3 entry into polarized human intestinal cells (Caco-2) occurs by a caveolin-dependent but dynamin-independent mechanism that requires DAF-mediated tyrosine kinase signals. In this study, we examined how CVB enter and infect nonpolarized HeLa cells and how DAF binding affects these processes. Using immunofluorescence microscopy and a combination of dominant-negative proteins, small interfering RNAs, and drugs targeting specific endocytic pathways, we found that both DAF-binding and non-DAF-binding virus isolates require dynamin and lipid rafts to enter and infect cells. Unlike what we observed in Caco-2 cells, CVB3 entered HeLa cells with CAR. We found no role for clathrin, endosomal acidification, or caveolin. Inhibition of tyrosine kinases blocked an early event in infection but did not prevent entry of virus into the cell. These results indicate that CVB3 entry into nonpolarized HeLa cells differs significantly from entry into polarized Caco-2 cells and is not influenced by virus binding to DAF.


Assuntos
Antígenos CD55/metabolismo , Dinaminas/metabolismo , Enterovirus Humano B/metabolismo , Microdomínios da Membrana/metabolismo , Internalização do Vírus , Animais , Benzamidas , Antígenos CD55/genética , Células CACO-2 , Caveolinas/metabolismo , Infecções por Coxsackievirus/metabolismo , Filipina/metabolismo , Células HeLa , Humanos , Mesilato de Imatinib , Piperazinas/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Pirimidinas/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo
17.
Cell Host Microbe ; 6(1): 91-8, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19616768

RESUMO

In cultured cells, infection by group B coxsackievirus (CVB) is mediated by the coxsackievirus and adenovirus receptor (CAR), but the importance of this molecule in CVB-induced disease has not been determined. We generated mice with tissue-specific ablation of CAR within each of two major CVB target organs, the pancreas and heart. In the pancreas, deletion of CAR resulted in a significant reduction in both virus titers and virus-induced tissue damage. Similarly, cardiomyocyte-specific CAR deletion resulted in a marked reduction in virus titer, infection-associated cytokine production, and histopathology within the heart. Consistent with the in vivo phenotype, CAR-deficient cardiomyocytes resisted infection in vitro. These results demonstrate a critical function for CAR in the pathogenesis of CVB infection in vivo and in virus tropism for the heart and pancreas.


Assuntos
Infecções por Coxsackievirus/prevenção & controle , Deleção de Genes , Miocardite/prevenção & controle , Pancreatite/prevenção & controle , Receptores Citoplasmáticos e Nucleares/genética , Animais , Células Cultivadas , Receptor Constitutivo de Androstano , Coração/virologia , Camundongos , Camundongos Knockout , Miocardite/virologia , Miocárdio/patologia , Pâncreas/patologia , Pâncreas/virologia , Pancreatite/virologia
19.
Cell Host Microbe ; 5(6): 517-21, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19527879

RESUMO

Most viruses infect their hosts by crossing the mucosal surfaces of the respiratory, gastrointestinal, or genital tracts, then spread--often through the bloodstream--to other organs; they are shed in bodily secretions to reach new hosts. At each stage in the cycle of infection viruses surmount significant anatomic barriers. This Minireview focuses on the role of intercellular junctions as barriers to virus dissemination, and the somewhat paradoxical observation that several viruses, rather than evading these barriers, target them directly by using junctional proteins as receptors.


Assuntos
Junções Intercelulares/fisiologia , Receptores Virais , Ligação Viral , Internalização do Vírus , Fenômenos Fisiológicos Virais , Modelos Biológicos
20.
J Virol ; 83(11): 5567-73, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19279092

RESUMO

A major obstacle to the use of adenovirus vectors derived from common human serotypes, such as human adenovirus 5 (AdHu5), is the high prevalence of virus-neutralizing antibodies in the human population. We previously constructed a variant of chimpanzee adenovirus 68 (AdC68) that maintained the fundamental properties of the carrier but was serologically distinct from AdC68 and resisted neutralization by AdC68 antibodies. In the present study, we tested whether this modified vector, termed AdCDQ, could induce transgene product-specific CD8(+) T cells in mice with preexisting neutralizing antibody to wild-type AdC68. Contrary to our expectation, the data show conclusively that antibodies that fail to neutralize the AdCDQ mutant vector in vitro nevertheless impair the vector's capacity to transduce cells and to stimulate a transgene product-specific CD8(+) T-cell response in vivo. The results thus suggest that in vitro neutralization assays may not reliably predict the effects of virus-specific antibodies on adenovirus vectors in vivo.


Assuntos
Adenoviridae/genética , Adenoviridae/imunologia , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/farmacologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Vacinas Virais/imunologia , Animais , Especificidade de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Feminino , Genes Reporter/genética , Imunoensaio , Camundongos , Camundongos Endogâmicos BALB C , Receptores Fc/imunologia , Transgenes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...