Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
PLoS Pathog ; 20(2): e1012022, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38359079

RESUMEN

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.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Humanos , Enterovirus/metabolismo , Enterovirus Humano A/genética , Enterovirus Humano A/metabolismo , Membrana Celular/metabolismo , Línea Celular , Receptores Depuradores/genética , Receptores Depuradores/metabolismo , Proteínas de Membrana de los Lisosomas/genética
2.
J Neurosci ; 36(37): 9558-71, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27629708

RESUMEN

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.


Asunto(s)
Enfermedad de Alzheimer/patología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/deficiencia , Hipocampo/patología , Neurogénesis/genética , Plasticidad Neuronal/genética , Sinapsis/metabolismo , Factores de Edad , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Animales , Células Cultivadas , Trastornos del Conocimiento/etiología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos , Potenciales Postsinápticos Excitadores/genética , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Nestina/genética , Nestina/metabolismo
3.
J Virol ; 90(12): 5601-5610, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27030267

RESUMEN

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.


Asunto(s)
Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/química , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/metabolismo , Enterovirus Humano B/fisiología , Acoplamiento Viral , Animales , Células CHO , Pollos , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/genética , Cricetulus , Enterovirus Humano B/química , Glicosilación , Células HeLa , Humanos , Dominios de Inmunoglobulinas/genética , Mutación , Replicación Viral
4.
PLoS Pathog ; 11(10): e1005184, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26430888

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Bencenosulfonatos/farmacología , Infecciones por Enterovirus/virología , Heparitina Sulfato/metabolismo , Glicoproteínas de Membrana/metabolismo , Acoplamiento Viral/efectos de los fármacos , Sitios de Unión , Cápside/química , Cápside/efectos de los fármacos , Cápside/metabolismo , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/metabolismo , Infecciones por Enterovirus/metabolismo , Células HeLa , Humanos , Células Jurkat , Modelos Moleculares , Datos de Secuencia Molecular , Suramina/análogos & derivados
5.
J Virol ; 89(2): 1324-8, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25392210

RESUMEN

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.


Asunto(s)
Antígenos CD55/metabolismo , Proteínas de la Cápside/metabolismo , Enterovirus Humano B/fisiología , Interacciones Huésped-Patógeno , Receptores Virales/metabolismo , Acoplamiento Viral , Secuencia de Aminoácidos , Animales , Antígenos CD55/genética , Línea Celular , Análisis Mutacional de ADN , Humanos , Ratones , Datos de Secuencia Molecular , Receptores Virales/genética
6.
J Virol ; 89(8): 4311-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25653430

RESUMEN

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.


Asunto(s)
Antígenos CD55/metabolismo , Susceptibilidad a Enfermedades/virología , Enterovirus Humano B/fisiología , Infecciones por Enterovirus/metabolismo , Mucosa Intestinal/metabolismo , Animales , Cartilla de ADN/genética , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Transgénicos , Microscopía Electrónica , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Virol ; 88(1): 434-43, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24155402

RESUMEN

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.


Asunto(s)
Autofagia , Enterovirus Humano B/fisiología , Mucosa Intestinal/virología , Fusión de Membrana/fisiología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Relacionadas con la Autofagia , Beclina-1 , Células CACO-2 , Endosomas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
8.
PLoS Pathog ; 9(7): e1003511, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23935488

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , Enterovirus Humano A/inmunología , Leucocitos/inmunología , Glicoproteínas de Membrana/metabolismo , Proteínas Virales de Fusión/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Sitios de Unión , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Secuencia Conservada , Enterovirus Humano A/fisiología , Interacciones Huésped-Patógeno , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Células Jurkat , Leucocitos/metabolismo , Leucocitos/virología , Lisina/química , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Tirosina/análogos & derivados , Tirosina/química , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Acoplamiento Viral
9.
J Virol ; 87(16): 8884-95, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740983

RESUMEN

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.


Asunto(s)
Colesterol/metabolismo , Dinaminas/metabolismo , Enterovirus Humano B/fisiología , Células Epiteliales/fisiología , Interacciones Huésped-Patógeno , Pinocitosis , Internalización del Virus , Oxidorreductasas de Alcohol/metabolismo , Células CACO-2 , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/virología , Humanos , Proteína Quinasa C/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Vesículas Transportadoras/virología
10.
J Virol ; 86(23): 12571-81, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22973031

RESUMEN

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.


Asunto(s)
Antígenos CD55/química , Enterovirus Humano B/ultraestructura , Modelos Moleculares , Conformación Proteica , Receptores Virales/química , Antígenos CD55/metabolismo , Línea Celular Tumoral , Microscopía por Crioelectrón , Cristalización , Cristalografía por Rayos X , Humanos , Receptores Virales/metabolismo
11.
Adv Exp Med Biol ; 790: 24-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23884584

RESUMEN

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.


Asunto(s)
Picornaviridae/fisiología , Internalización del Virus , Animales , Antígenos CD55/fisiología , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus/fisiología , Endocitosis , Humanos , Picornaviridae/ultraestructura , Receptores Virales/fisiología , Acoplamiento Viral
12.
J Virol ; 85(14): 7436-43, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21561916

RESUMEN

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.


Asunto(s)
Sustitución de Aminoácidos , Cápside , Enterovirus Humano B/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Cartilla de ADN , Enterovirus Humano B/química , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
13.
J Virol ; 83(21): 11064-77, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19710132

RESUMEN

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.


Asunto(s)
Antígenos CD55/metabolismo , Dinaminas/metabolismo , Enterovirus Humano B/metabolismo , Microdominios de Membrana/metabolismo , Internalización del Virus , Animales , Benzamidas , Antígenos CD55/genética , Células CACO-2 , Caveolinas/metabolismo , Infecciones por Coxsackievirus/metabolismo , Filipina/metabolismo , Células HeLa , Humanos , Mesilato de Imatinib , Piperazinas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Pirimidinas/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo
14.
J Virol ; 83(11): 5567-73, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19279092

RESUMEN

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.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vacunas Virales/inmunología , Animales , Especificidad de Anticuerpos , Linfocitos T CD8-positivos/inmunología , Femenino , Genes Reporteros/genética , Inmunoensayo , Ratones , Ratones Endogámicos BALB C , Receptores Fc/inmunología , Transgenes/genética
15.
Trends Microbiol ; 16(2): 44-7, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18191571

RESUMEN

To initiate infection, poliovirus must release its RNA genome into the cytoplasm of a target cell, a process called 'uncoating'. How this occurs has remained uncertain, despite studies over several decades. Two new studies re-address the question of poliovirus entry. The results suggest that poliovirus enters different cells by different mechanisms, and point to a role for virus-induced intracellular signals in the process.


Asunto(s)
Colorantes Fluorescentes/metabolismo , Poliovirus/fisiología , Internalización del Virus , Caveolas/virología , Vesículas Cubiertas por Clatrina/virología , Citoplasma/virología , Endocitosis , Células Endoteliales/virología , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Poliovirus/ultraestructura , ARN Viral/metabolismo , Receptores Virales/metabolismo , Coloración y Etiquetado , Virión/metabolismo
16.
Circ Res ; 98(7): 923-30, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16543498

RESUMEN

The coxsackievirus and adenovirus receptor (CAR), which mediates infection by the viruses most commonly associated with myocarditis, is a transmembrane component of specialized intercellular junctions, including the myocardial intercalated disc; it is known to mediate cell-cell recognition, but its natural function is poorly understood. We used conditional gene targeting to investigate the possible functions of CAR during embryonic development, generating mice with both germline and tissue-specific defects in CAR expression. Homozygous germline deletion of CAR exon 2 or cardiomyocyte-specific gene deletion at embryonic day 10 (E10) mediated by Cre recombinase expressed under the control of the cardiac troponin T promoter resulted in death by E12.5; embryos showed marked cardiac abnormalities by E10.5, with hyperplasia of the left ventricular myocardium, distention of the cardinal veins, and abnormalities of sinuatrial valves. Within the hyperplastic left ventricle, increased numbers of proliferating cells were evident; persistent expression of N-myc in the hyperplastic myocardium and attenuated expression of the trabecular markers atrial natriuretic factor and bone morphogenic protein 10 indicated that proliferating cardiomyocytes had failed to differentiate and form normal trabeculae. In electron micrographs, individual CAR-deficient cardiomyocytes within the left ventricle appeared normal, but intercellular junctions were ill-formed or absent, consistent with the known function of CAR as a junctional molecule; myofibrils were also poorly organized. When cardiomyocyte-specific deletion occurred somewhat later (by E11, mediated by Cre under control of the alpha-myosin heavy chain promoter), animals survived to adulthood and did not have evident cardiac abnormalities. These results indicate that during a specific temporal window, CAR expression on cardiomyocytes is essential for normal cardiac development. In addition, the results suggest that CAR-mediated intercellular contacts may regulate proliferation and differentiation of cardiomyocytes within the embryonic left ventricular wall.


Asunto(s)
Cardiopatías Congénitas/genética , Hipertrofia Ventricular Izquierda/genética , Receptores Virales/deficiencia , Receptores Virales/genética , Nodo Sinoatrial/anomalías , Animales , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Cartilla de ADN , Exones , Femenino , Eliminación de Gen , Mutación de Línea Germinal , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa
17.
Curr Opin Microbiol ; 6(4): 386-91, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12941410

RESUMEN

Viruses attach to specific receptors, but identified receptors are often absent from the epithelial surfaces where infections begin. Viruses can bind to alternative receptor molecules present on epithelial surfaces and they can enter hosts in ways that bypass mucosal barriers to infection. To understand how viruses cross the mucosa to initiate infection we need new information about the mechanisms of attachment and entry into cells, but we also need in vivo studies to define precisely where infection occurs.


Asunto(s)
Membrana Mucosa/virología , Receptores de Superficie Celular/fisiología , Virosis/fisiopatología , Virus/patogenicidad , Animales , Humanos , Virulencia , Virosis/transmisión
18.
Adv Drug Deliv Rev ; 57(6): 869-82, 2005 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-15820557

RESUMEN

The coxsackievirus and adenovirus receptor (CAR) mediates cell attachment and infection by coxsackie B viruses and by a number of adenoviruses. CAR also mediates homotypic intercellular interactions. In polarized epithelial cells, CAR is closely associated with the tight junction, where it contributes to the barrier to paracellular flow of solutes and macromolecules. CAR's biological roles are not well defined, but emerging evidence suggests that it may function during embryonic development and in regulating cell proliferation.


Asunto(s)
Receptores Virales/química , Receptores Virales/fisiología , Uniones Estrechas/virología , Secuencia de Aminoácidos , Animales , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Humanos , Datos de Secuencia Molecular , Receptores Virales/genética , Uniones Estrechas/química , Uniones Estrechas/genética
19.
Clin Cancer Res ; 8(6): 1915-23, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12060636

RESUMEN

PURPOSE: Several barriers that collectively restrict gene delivery by viral vectors in vivo have been described. Previously, we identified soluble chondroitin sulfate-proteoglycans/glycosaminoglycans in malignant pleural effusions (MPEs) as inhibitors of retroviral vector transduction. Soluble components of MPE also inhibited adenoviral (Ad) gene transfer, and the factors were characteristically filterable, titrable, stable at 56 degrees C, and blocked the binding of Ad to target cells. Depleting immunoglobulin from MPE, partially reversed the block to Ad transduction, instigating a search for additional factors that bound Ad in MPE. EXPERIMENTAL DESIGN: Vector-protein interactions were identified after the resolution of MPE-components by SDS-PAGE. Viral overlays and immunoblots delineated significant interactions, and the potential relevance of those interactions was tested in transduction efficiency bioassays. RESULTS: Immunoglobulin is the predominant factor inhibiting Ad gene transfer in MPE. Albumin also interacted with Ad, although at predicted serum concentrations, it did not effect Ad transduction efficiency in vitro. Soluble coxsackievirus-Ad receptor (sCAR) was then identified in MPE. In a survey of 18 MPE, the mean concentration of sCAR was variable and estimated to be 3.51 +/- 5.02 ng/ml by ELISA. The impact of sCAR on transduction efficiency in this milieu was next assessed. Whereas immunodepletion of sCAR from MPE by affinity chromatography resulted in enhanced gene transfer within MPE, the inhibition of adenoviral gene transfer was not evident when the predicted concentrations of recombinant sCAR were added into the transduction medium. CONCLUSIONS: These studies indicate that, in addition to anti-Ad antibodies, other specific and nonspecific factors interact with viral vectors and may impair gene transfer in the tumor milieu. The presence of sCAR in MPE puts forward the notion that in certain contexts (e.g., within the extracellular matrix of solid tumors) the concentrations of secreted (or shed) CAR may be high enough to effectively compete with Ad gene delivery.


Asunto(s)
Adenoviridae/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Enterovirus/genética , Técnicas de Transferencia de Gen , Neoplasias Pulmonares/metabolismo , Receptores Virales/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Electroforesis en Gel de Poliacrilamida , Ensayo de Inmunoadsorción Enzimática , Expresión Génica , Terapia Genética , Vectores Genéticos , Humanos , Operón Lac/fisiología , Neoplasias Pulmonares/genética , Ratones , Derrame Pleural/metabolismo , Receptores Virales/inmunología , Células Tumorales Cultivadas
20.
Cell Host Microbe ; 18(2): 221-32, 2015 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-26269957

RESUMEN

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.


Asunto(s)
Autofagia , Cisteína Endopeptidasas/metabolismo , Enterovirus Humano B/fisiología , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Proteínas Virales/metabolismo , Replicación Viral , Proteasas Virales 3C , Células CACO-2 , Enterovirus Humano B/metabolismo , Silenciador del Gen , Pruebas Genéticas , Humanos , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA