Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 8: 567, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28572804

RESUMEN

Hantavirus infection is an acute zoonosis that clinically manifests in two primary forms, hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). HFRS is endemic in Europe and Russia, where the mild form of the disease is prevalent in the Tatarstan region. HPS is endemic in Argentina, as well as other countries of North and South American. HFRS and HPS are usually acquired via the upper respiratory tract by inhalation of virus-contaminated aerosol. Although the pathogenesis of HFRS and HPS remains largely unknown, postmortem tissue studies have identified endothelial cells as the primary target of infection. Importantly, cell damage due to virus replication, or subsequent tissue repair, has not been documented. Since no single factor has been identified that explains the complexity of HFRS or HPS pathogenesis, it has been suggested that a cytokine storm may play a crucial role in the manifestation of both diseases. In order to identify potential serological markers that distinguish HFRS and HPS, serum samples collected during early and late phases of the disease were analyzed for 48 analytes using multiplex magnetic bead-based assays. Overall, serum cytokine profiles associated with HPS revealed a more pro-inflammatory milieu as compared to HFRS. Furthermore, HPS was strictly characterized by the upregulation of cytokine levels, in contrast to HFRS where cases were distinguished by a dichotomy in serum cytokine levels. The severe form of hantavirus zoonosis, HPS, was characterized by the upregulation of a higher number of cytokines than HFRS (40 vs 21). In general, our analysis indicates that, although HPS and HFRS share many characteristic features, there are distinct cytokine profiles for these diseases. These profiles suggest a strong activation of an innate immune and inflammatory responses are associated with HPS, relative to HFRS, as well as a robust activation of Th1-type immune responses. Finally, the results of our analysis suggest that serum cytokines profiles of HPS and HFRS cases are consistent with the presence of extracellular matrix degradation, increased mononuclear leukocyte proliferation, and transendothelial migration.

2.
Front Microbiol ; 7: 1077, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27486439

RESUMEN

Hantavirus infection is associated with two frequently fatal diseases in humans: Hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). The pathogenesis of hantavirus infection is complex and not fully understood; however, it is believed to involve virus-induced hyperinflammatory immune responses. Thrombospondin-1 (THBS1) is a large homotrimeric protein that plays a putative role in regulating blood homeostasis. Hyperresponsiveness to inflammatory stimuli has also been associated with defects in the THBS1 gene. Our data suggest that hantavirus infection of human umbilical cord vein endothelial cells (HUVEC) suppress the accumulation of THBS1 in the extracellular matrix. Additionally, this suppression is dependent on virus replication, implying a direct mechanism of action. Our data also imply that the pathogenic Andes and Hantaan strains inhibit THBS1 expression while the non-pathogenic Prospect Hill strain showed little inhibition. These observations suggest that a dysregulation of THBS1 may contribute to the pathogenesis of hantavirus infection.

3.
Arch Virol ; 156(10): 1847-51, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21625978

RESUMEN

Human cytomegalovirus (HCMV) latency is poorly understood. We previously described a novel HCMV latency-associated transcript, UL81-82ast, coding for a protein designated LUNA (latency unique natural antigen). The aim of this study was to confirm the presence of LUNA in HCMV-seropositive donors. Standard co-immunoprecipitation and ELISA assays were used to detect antibodies against the LUNA protein in the sera of HCMV-seropositive donors. Specific antibodies against LUNA were detected in all HCMV-seropositive donors but in none of the seronegative donors. These data confirm that LUNA is expressed during in vivo infections and is capable of eliciting an immune response.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/genética , Regulación Viral de la Expresión Génica , Proteínas Virales/genética , Anticuerpos Antivirales/inmunología , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/inmunología , Humanos , Proteínas Virales/inmunología , Latencia del Virus
4.
J Biol Chem ; 284(13): 8654-60, 2009 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-19179334

RESUMEN

Hantaviruses are distributed worldwide and can cause a hemorrhagic fever or a cardiopulmonary syndrome in humans. Mature virions consist of RNA genome, nucleocapsid protein, RNA polymerase, and two transmembrane glycoproteins, G1 and G2. The ectodomain of G1 is surface-exposed; however, it has a 142-residue C-terminal cytoplasmic tail that plays important roles in viral assembly and host-pathogen interaction. Here we show by NMR, circular dichroism spectroscopy, and mutagenesis that a highly conserved cysteine/histidine-rich region in the G1 tail of hantaviruses forms two CCHC-type classical zinc fingers. Unlike classical zinc fingers, however, the two G1 zinc fingers are intimately joined together, forming a compact domain with a unique fold. We discuss the implication of the hantaviral G1 zinc fingers in viral assembly and host-pathogen interaction.


Asunto(s)
Orthohantavirus/química , Proteínas del Envoltorio Viral/química , Membrana Celular/química , Membrana Celular/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Orthohantavirus/fisiología , Síndrome Pulmonar por Hantavirus/metabolismo , Fiebres Hemorrágicas Virales/metabolismo , Interacciones Huésped-Patógeno/fisiología , Humanos , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína/fisiología , Proteínas del Envoltorio Viral/metabolismo , Ensamble de Virus/fisiología , Dedos de Zinc/fisiología
5.
J Biol Chem ; 283(42): 28297-304, 2008 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-18687679

RESUMEN

The hantaviruses are emerging infectious viruses that in humans can cause a cardiopulmonary syndrome or a hemorrhagic fever with renal syndrome. The nucleocapsid (N) is the most abundant viral protein, and during viral assembly, the N protein forms trimers and packages the viral RNA genome. Here, we report the NMR structure of the N-terminal domain (residues 1-74, called N1-74) of the Andes hantavirus N protein. N1-74 forms two long helices (alpha1 and alpha2) that intertwine into a coiled coil domain. The conserved hydrophobic residues at the helix alpha1-alpha2 interface stabilize the coiled coil; however, there are many conserved surface residues whose function is not known. Site-directed mutagenesis, CD spectroscopy, and immunocytochemistry reveal that a point mutation in the conserved basic surface formed by Arg22 or Lys26 lead to antibody recognition based on the subcellular localization of the N protein. Thus, Arg22 and Lys26 are likely involved in a conformational change or molecular recognition when the N protein is trafficked from the cytoplasm to the Golgi, the site of viral assembly and maturation.


Asunto(s)
Proteínas de la Nucleocápside/química , Orthohantavirus/metabolismo , Animales , Arginina/química , Células COS , Chlorocebus aethiops , Dicroismo Circular , Citoplasma/metabolismo , Aparato de Golgi/metabolismo , Lisina/química , Espectroscopía de Resonancia Magnética , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
6.
J Wildl Dis ; 42(4): 819-24, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17255449

RESUMEN

We examined the impact of season and habitat on Sin Nombre virus (SNV) seroprevalence in deer mice (Peromyscus maniculatus) in Utah's Great Basin Desert from May 2002 through summer 2003. Low mouse captures in 2002 limited analysis for that year. In two seasons during 2003, mouse density and sagebrush cover were positively linked (spring: r = 0.8, P = 0.01; summer: r = 0.8, P = 0.04). In the spring, seroprevalence was negatively correlated with density (r = -0.9, P< 0.01); male and female antibody prevalence did not differ; and scarring was unrelated to antibody status. In the summer, density and antibody prevalence were unrelated; male seroprevalence was higher (chi(2) = 3.6, P = 0.05); and seropositive mice had more scars (t = 2.5, P = 0.02). We speculate nesting behavior could maintain SNV over the winter, whereas summer territoriality could be responsible for transmission.


Asunto(s)
Anticuerpos Antivirales/sangre , Síndrome Pulmonar por Hantavirus/veterinaria , Peromyscus , Enfermedades de los Roedores/epidemiología , Virus Sin Nombre/inmunología , Animales , Ecosistema , Femenino , Síndrome Pulmonar por Hantavirus/epidemiología , Síndrome Pulmonar por Hantavirus/transmisión , Masculino , Peromyscus/virología , Densidad de Población , Enfermedades de los Roedores/transmisión , Estaciones del Año , Estudios Seroepidemiológicos , Factores Sexuales , Utah/epidemiología
7.
Vaccine ; 24(3): 327-34, 2006 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-16125285

RESUMEN

Previously, we have shown that CMV isolated from deer mouse could be used in vivo and in vitro to express Sin Nombre virus (SNV) glycoprotein G1 in deer mice [Rizvanov AA, van Geelen AG, Morzunov S, et al. Generation of a recombinant cytomegalovirus for expression of a hantavirus glycoprotein. J.Virol. 2003;77(22):12203-10]. In this report, we further characterize replication of wild-type (wt) and recombinant Peromyscus CMV (PCMV) in vivo and in vitro using realtime PCR, and infectious center assays. Our findings indicate that both wt PCMV and recombinant PCMV establish persistent infections in P. maniculatus. In addition, we demonstrated that gamma irradiation of PCMV infected mice resulted in reactivation of recombinant PCMV in persistently infected mice.


Asunto(s)
Citomegalovirus/inmunología , Orthohantavirus/genética , Orthohantavirus/inmunología , Peromyscus/virología , Proteínas del Envoltorio Viral/biosíntesis , Vacunas Virales/inmunología , Animales , Citomegalovirus/genética , ADN/biosíntesis , ADN/genética , Replicación del ADN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vacunas Sintéticas/inmunología , Proteínas del Envoltorio Viral/genética , Replicación Viral , Irradiación Corporal Total
8.
Infect Immun ; 74(1): 362-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16368991

RESUMEN

Mannan is a major cell wall component found in Candida species. Natural antimannan antibody is present in sera from most normal adults, but its role in host resistance to hematogenously disseminated candidiasis is unknown. The purpose of this study was to develop recombinant human antimannan antibody and to study its protective function. A phage Fab display combinatorial library containing Fab genes from bone marrow lymphocytes was screened with Candida albicans yeast cells and chemically purified mannan. One antimannan Fab, termed M1, was converted to a full-length immunoglobulin G1 antibody, M1g1, and M1g1 was produced in CHO cells. The M1g1 epitope was found in C. albicans serotypes A and B, Candida tropicalis, Candida guilliermondii, Candida glabrata, and Candida parapsilosis. Its expression was active at both 23 degrees C and 37 degrees C and uniform over the cell surface. BALB/c mice passively immunized with M1g1 were more resistant than control mice to a lethal hematogenous infection by C. albicans, as evidenced by extension of survival in an M1g1 dose-dependent manner (P, 0.08 to <0.001) and by reduction in number of infection foci and their size in the kidney. In vitro studies found that M1g1 promoted phagocytosis and phagocytic killing of C. albicans yeast cells by mouse peritoneal macrophages and was required for activation of the mouse complement cascade. Thus, human antimannan antibody may have a protective role in host resistance to systemic candidiasis.


Asunto(s)
Candidiasis/inmunología , Inmunoglobulina G/fisiología , Mananos/inmunología , Animales , Células CHO , Candidiasis/metabolismo , Proteínas del Sistema Complemento/metabolismo , Cricetinae , Cricetulus , Femenino , Humanos , Inmunidad Innata/genética , Fragmentos Fab de Inmunoglobulinas/genética , Inmunoglobulina G/genética , Riñón/microbiología , Riñón/patología , Ratones , Ratones Endogámicos BALB C , Fagocitosis/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico
9.
Curr Mol Med ; 5(8): 773-90, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16375712

RESUMEN

Hantaviruses are tri-segmented negative sense single stranded RNA viruses that belong to the family Bunyaviridae. In nature, hantaviruses are exclusively maintained in the populations of their specific rodent hosts. In their natural host species, hantaviruses usually develop a persistent infection with prolonged virus shedding in excreta. Humans become infected by inhaling virus contaminated aerosol. Unlike asymptomatic infection in rodents, hantaviruses cause two acute febrile diseases in humans: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). The mortality rate varies from 0.1% to 40% depending on the virus involved. Hantaviruses are distributed world wide, with over 150,000 HFRS and HPS cases being registered annually. In this review we summarize current knowledge on hantavirus molecular biology, epidemiology, genetic diversity and co-evolution with rodent hosts. In addition, special attention was given in this review to describing clinical manifestation of HFRS and HPS, and advances in our current understanding of the host immune response, treatment, and prevention.


Asunto(s)
Infecciones por Hantavirus/inmunología , Infecciones por Hantavirus/virología , Orthohantavirus/genética , Orthohantavirus/patogenicidad , Animales , Citocinas/inmunología , Evolución Molecular , Genoma Viral , Orthohantavirus/metabolismo , Infecciones por Hantavirus/terapia , Humanos , Inmunidad
10.
Virology ; 331(2): 307-15, 2005 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-15629773

RESUMEN

This study was designed to investigate the trafficking of Andes virus (ANDV) and Sin Nombre virus (SNV) glycoproteins and to determine if ANDV or SNV glycoproteins G1 and G2 could be substituted for each other while still retaining normal trafficking. Trafficking of Hantaan virus (HNTV) and SNV glycoproteins has been studied and conflicting results were published regarding the Golgi targeting of G1 and G2 when expressed individually. The results reported in this manuscript suggest that both SNV and ANDV G1 and G2 expressed together, either from a single glycoprotein precursor (GPC) or from separate cDNAs, co-localize to the Golgi complex (GC). When expressed individually, neither G1 nor G2 was able to translocate from the endoplasmic reticulum (ER) to the GC. Interestingly, when ANDV G1 and SNV G2 or ANDV G2 and SNV G1 are co-expressed, they interact and are colocalized in the GC.


Asunto(s)
Regulación Viral de la Expresión Génica , Orthohantavirus/metabolismo , Virus Sin Nombre/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Células HeLa , Humanos , Fracciones Subcelulares/metabolismo , Células Vero , Proteínas del Envoltorio Viral/análisis , Proteínas del Envoltorio Viral/genética
11.
J Med Virol ; 75(2): 267-75, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15602733

RESUMEN

MxA is a cellular protein activated in cells treated with type I interferons. MxA protein is a member of the dynamin superfamily of large guanosine triphosphatases. A unique property of Mx1 GTPases is their antiviral activity against a wide range of RNA viruses. Replication of several hantavirus strains is inhibited in cells constitutively expressing MxA protein. We have found that Andes virus (ANDV) infection up regulates transcription of MxA RNA and expression of MxA protein in human endothelial cells in vitro. Activation of MxA gene expression requires virus replication. Up-regulation of MxA gene expression in hantavirus infected cells varies depending on the cell type. The degree of activation of MxA gene expression is inversely correlated with the efficiency of hantavirus replication. Additionally, MxA protein is co-localized with hantavirus nucleocapsid protein in infected cell. Our data suggest that MxA by interacting with the virus nucleocapsid protein inhibits production of new infectious virus particles.


Asunto(s)
Células Endoteliales/inmunología , Proteínas de Unión al GTP/inmunología , Regulación de la Expresión Génica/inmunología , Orthohantavirus/inmunología , Animales , Línea Celular , Chlorocebus aethiops , Células Endoteliales/metabolismo , Proteínas de Unión al GTP/biosíntesis , Infecciones por Hantavirus/inmunología , Infecciones por Hantavirus/metabolismo , Humanos , Proteínas de Resistencia a Mixovirus , Regulación hacia Arriba , Células Vero , Replicación Viral/inmunología
12.
Pediatr Infect Dis J ; 23(10): 974-5, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15602208

RESUMEN

Occasionally a case report can provide enormous insight into the pathogenesis of an infectious disease. In this instance, 2 Iowa boys were bitten by the same mouse on May 1, 1999. Three weeks later, both boys developed sin nombre hantavirus pulmonary syndrome. Thus, the incubation period of hantavirus infection has been documented. Both survived; one of the 2 boys continued to have a very high titer of hantavirus antibody 4 years after the mouse bite.


Asunto(s)
Infecciones por Hantavirus , Síndrome Pulmonar por Hantavirus , Adolescente , Animales , Anticuerpos Antivirales/sangre , Mordeduras y Picaduras/complicaciones , Niño , Reservorios de Enfermedades , Orthohantavirus/inmunología , Humanos , Masculino , Ratones , Factores de Tiempo
13.
J Virol ; 77(22): 12203-10, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14581557

RESUMEN

A cytomegalovirus (CMV) was isolated from its natural host, Peromyscus maniculatus, and was designated Peromyscus CMV (PCMV). A recombinant PCMV was constructed that contained Sin Nombre virus glycoprotein G1 (SNV-G1) fused in frame to the enhanced green fluorescent protein (EGFP) gene inserted into a site homologous to the human CMV UL33 (P33) gene. The recombinant CMV was used for expression and immunization of deer mice against SNV-G1. The results of the study indicate that P. maniculatus could be infected with as few as 10 virus particles of recombinant virus. Challenge of P. maniculatus with either recombinant or wild-type PCMV produced no overt pathology in infected animals. P. maniculatus immunized with recombinant virus developed an antibody response to SNV and EGFP. When rechallenged with recombinant virus, animals exhibited an anamnestic response against SNV. Interestingly, a preexisting immune response against PCMV did not prevent reinfection with recombinant PCMV.


Asunto(s)
Citomegalovirus/genética , Glicoproteínas/biosíntesis , Peromyscus/virología , Proteínas Recombinantes/biosíntesis , Virus Sin Nombre/inmunología , Vacunas Sintéticas/inmunología , Proteínas Virales/biosíntesis , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Citomegalovirus/inmunología , ADN Polimerasa Dirigida por ADN/análisis , Inmunización , Datos de Secuencia Molecular , Receptores de Quimiocina/análisis , Virus Sin Nombre/química , Proteínas Virales/análisis
14.
Am J Trop Med Hyg ; 67(3): 310-8, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12408674

RESUMEN

Many researchers have speculated that infection dynamics of Sin Nombre virus are driven by density patterns of its major host, Peromyscus maniculatus. Few, if any, studies have examined this question systematically at a realistically large spatial scale, however. We collected data from 159 independent field sites within a 1 million-hectare study area in Nevada and California, from 1995-1998. In 1997, there was a widespread and substantial reduction in host density. This reduction in host density did not reduce seroprevalence of antibody to Sin Nombre virus within host populations. During this period, however, there was a significant reduction in the likelihood that antibody-positive mice had detectable virus in their blood, as determined by reverse-transcriptase polymerase chain reaction. Our findings suggest 2 possible causal mechanisms for this reduction: an apparent change in the age structure of host populations and landscape-scale patterns of host density. This study indicates that a relationship does exist between host density and infection dynamics and that this relationship concurrently operates at different spatial scales. It also highlights the limitations of antibody seroprevalence as a metric of infections, especially during transient host-density fluctuations.


Asunto(s)
Peromyscus/virología , Virus Sin Nombre/patogenicidad , Animales , Antígenos Virales/sangre , Ensayo de Inmunoadsorción Enzimática , Dinámica Poblacional , ARN Viral/sangre , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virus Sin Nombre/genética , Virus Sin Nombre/inmunología
15.
Virus Res ; 89(1): 131-43, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12367756

RESUMEN

We have determined the genomic sequence of an Andes virus (ANDV) strain isolated from an infected Oligoryzomys longicaudatus rodent trapped in Chile in 1997. This strain, for which we propose the designation Chile R123, reproduces essential attributes of hantavirus pulmonary syndrome (HPS) when injected intramuscularly into laboratory hamsters (Hooper et al., Virology 289 (2001) 6-14). The L, M, and S segment sequences of Chile R123 are 6562, 3671, and 1871 nt long, respectively, with an overall G+C content of 38.5%. These respective genome segments could encode a 247 kd RNA-dependent RNA polymerase (RdRP), 126 kd glycoprotein precursor (GPC), and 48 kd nucleocapsid (N) protein, in line with other Sigmodontine rodent-associated hantaviruses. Among hantaviruses for which complete genomic sequences are available, Chile R123 is most closely related to Sin Nombre virus (SNV) strain NM R11, with greater than 85% amino acid identity between translated L and S segments and 78% amino acid identity between translated M segments. Because Chile R123 shares essentially 100% amino acid identity in regions of overlap with partially sequenced Argentinian and Chilean ANDV strains, Syrian hamster pathogenicity and the potential for interhuman transmission are features likely common to all ANDV strains.


Asunto(s)
Secuencia de Bases , Brotes de Enfermedades , Infecciones por Hantavirus/veterinaria , Orthohantavirus/química , Enfermedades de los Roedores/virología , Animales , Chile , Cricetinae , Modelos Animales de Enfermedad , Genoma Viral , Orthohantavirus/clasificación , Orthohantavirus/genética , Orthohantavirus/patogenicidad , Infecciones por Hantavirus/epidemiología , Infecciones por Hantavirus/fisiopatología , Infecciones por Hantavirus/virología , Humanos , Mesocricetus , Datos de Secuencia Molecular , Muridae/virología , Filogenia , Enfermedades de los Roedores/epidemiología , Análisis de Secuencia de ADN
16.
J Virol ; 76(8): 3765-73, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11907216

RESUMEN

Nucleotide sequences were determined for the complete S genome segments of the six distinct hantavirus genotypes from Argentina and for two cell culture-isolated Andes virus strains from Chile. Phylogenetic analysis indicates that, although divergent from each other, all Argentinian hantavirus genotypes group together and form a novel phylogenetic clade with the Andes virus. The previously characterized South American hantaviruses Laguna Negra virus and Rio Mamore virus make up another clade that originates from the same ancestral node as the Argentinian/Chilean viruses. Within the clade of Argentinian/Chilean viruses, three subclades can be defined, although the branching order is somewhat obscure. These are made of (i) "Lechiguanas-like" virus genotypes, (ii) Maciel virus and Pergamino virus genotypes, and (iii) strains of the Andes virus. Two hantavirus genotypes from Brazil, Araraquara and Castello dos Sonhos, were found to group with Maciel virus and Andes virus, respectively. The nucleocapsid protein amino acid sequence variability among the members of the Argentinian/Chilean clade does not exceed 5.8%. It is especially low (3.5%) among oryzomyine species-associated virus genotypes, suggesting recent divergence from the common ancestor. Interestingly, the Maciel and Pergamino viruses fit well with the rest of the clade although their hosts are akodontine rodents. Taken together, these data suggest that under conditions in which potential hosts display a high level of genetic diversity and are sympatric, host switching may play a prominent role in establishing hantavirus genetic diversity. However, cospeciation still remains the dominant factor in the evolution of hantaviruses.


Asunto(s)
Variación Genética , Infecciones por Hantavirus/epidemiología , Infecciones por Hantavirus/virología , Orthohantavirus/genética , Filogenia , Animales , Argentina/epidemiología , Chile/epidemiología , Orthohantavirus/clasificación , Humanos , Datos de Secuencia Molecular , Proteínas de la Nucleocápside/genética , Enfermedades de los Roedores/epidemiología , Enfermedades de los Roedores/virología , Roedores , Análisis de Secuencia de ADN
17.
Viral Immunol ; 15(4): 609-25, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12513931

RESUMEN

Two clinical syndromes are associated with hantavirus infection in humans: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). Autopsy findings typically reveal a common feature of increased permeability in microvascular beds, suggesting vascular endothelium is a prime target for virus infection. Endothelial cells are susceptible to hantavirus infection; however, virus does not cause cytopathic effects, to explain increased endothelium permeability. Therefore, immune mechanisms were suggested to play a crucial role in hantavirus pathogenesis. In this review, we summarize data on hantavirus-induced immune disturbances and discuss their implication in capillary leakage caused by hantavirus infection.


Asunto(s)
Endotelio Vascular/inmunología , Síndrome Pulmonar por Hantavirus/inmunología , Síndrome Pulmonar por Hantavirus/fisiopatología , Fiebre Hemorrágica con Síndrome Renal/inmunología , Fiebre Hemorrágica con Síndrome Renal/fisiopatología , Orthohantavirus/patogenicidad , Endotelio Vascular/citología , Endotelio Vascular/virología , Orthohantavirus/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA