Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Vet Microbiol ; 227: 69-77, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30473354

RESUMEN

A severe infectious disease characterized by nephritis, hepatitis and splenitis has attacked goslings around Shandong province in China since 2016. A novel chicken-origin avian orthoreovirus (ARV) was isolated with LMH cells from affected goslings named Reo/Goose/SDPY/1116/17 (SDPY-ARV) strain, and the infection was successfully reproduced experimentally. The ARV-SDPY full genome sequencing was conducted using Next-Generation Sequencing (NGS) technique on Illumina HiSeq platform. The complete genome of SDPY-ARV was 23,427 bp in length and consist of 10 dsRNA segments ranged from 1192 bp (S4) to 3958 bp (L1) which encoding 12 viral proteins. Genomic sequence analysis showed that the SDPY-ARV strain is in the same branch with broiler, pheasant-origin ARV isolates, and shares 51.8-96.2% of nucleotide identity of σC gene with them; while only 49.3-50.3% with waterfowl isolates. In addition, the occurrence of 10 segments genetic reassortment of SDPY strain is confirmed among the PA15511, the 1733 and the PA13649 strains from America. In conclusion, the causative agent of gosling hemorrhagic necrotic hepatitis and nephritis occurring in China is a novel chicken-origin goose orthoreovirus.


Asunto(s)
Gansos/virología , Genoma Viral , Hepatitis Viral Animal/etiología , Orthoreovirus Aviar/genética , Orthoreovirus Aviar/aislamiento & purificación , Enfermedades de las Aves de Corral/virología , Infecciones por Reoviridae/veterinaria , Factores de Edad , Animales , Pollos , China/epidemiología , Genómica , Hepatitis Viral Animal/epidemiología , Hepatitis Viral Animal/virología , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Hígado/patología , Hígado/virología , Necrosis/veterinaria , Necrosis/virología , Sistemas de Lectura Abierta , Orthoreovirus Aviar/fisiología , Filogenia , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/transmisión , Infecciones por Reoviridae/epidemiología , Infecciones por Reoviridae/transmisión , Infecciones por Reoviridae/virología , Análisis de Secuencia de ADN , Proteínas Virales/genética
2.
J Hepatol ; 68(4): 682-690, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29274730

RESUMEN

BACKGROUND & AIM: Virus-induced fulminant hepatitis is a major cause of acute liver failure. During acute viral hepatitis the impact of type I interferon (IFN-I) on myeloid cells, including liver-resident Kupffer cells (KC), is only partially understood. Herein, we dissected the impact of locally induced IFN-I responses on myeloid cell function and hepatocytes during acute liver inflammation. METHODS: Two different DNA-encoded viruses, vaccinia virus (VACV) and murine cytomegalovirus (MCMV), were studied. In vivo imaging was applied to visualize local IFN-ß induction and IFN-I receptor (IFNAR) triggering in VACV-infected reporter mice. Furthermore, mice with a cell type-selective IFNAR ablation were analyzed to dissect the role of IFNAR signaling in myeloid cells and hepatocytes. Experiments with Cx3cr1+/gfp mice revealed the origin of reconstituted KC. Finally, mixed bone marrow chimeric mice were studied to specifically analyze the effect of IFNAR triggering on liver infiltrating monocytes. RESULTS: VACV infection induced local IFN-ß responses, which lead to IFNAR signaling primarily within the liver. IFNAR triggering was needed to control the infection and prevent fulminant hepatitis. The severity of liver inflammation was independent of IFNAR triggering of hepatocytes, whereas IFNAR triggering of myeloid cells protected from excessive inflammation. Upon VACV or MCMV infection KC disappeared, whereas infiltrating monocytes differentiated to KC afterwards. During IFNAR triggering such replenished monocyte-derived KC comprised more IFNAR-deficient than -competent cells in mixed bone marrow chimeric mice, whereas after the decline of IFNAR triggering both subsets showed an even distribution. CONCLUSION: Upon VACV infection IFNAR triggering of myeloid cells, but not of hepatocytes, critically modulates acute viral hepatitis. During infection with DNA-encoded viruses IFNAR triggering of liver-infiltrating blood monocytes delays the development of monocyte-derived KC, pointing towards new therapeutic strategies for acute viral hepatitis. LAY SUMMARY: Viral infection can cause fulminant hepatitis, which in turn is a major cause of acute liver failure. Herein, we aimed to study the role of type 1 interferon responses in acute viral hepatitis. We identified that during infection with DNA-encoded viruses, type 1 interferon receptor triggering of blood monocytes delays the development of monocyte-derived Kupffer cells. This points to new therapeutic strategies for acute viral hepatitis.


Asunto(s)
Hepatitis Viral Animal/fisiopatología , Macrófagos del Hígado/fisiología , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/fisiología , Enfermedad Aguda , Animales , Hepatitis Viral Animal/etiología , Ratones , Ratones Endogámicos C57BL , Vaccinia/fisiopatología
3.
J Hepatol ; 68(4): 733-743, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29154963

RESUMEN

BACKGROUND & AIMS: The macrophage scavenger receptor 1 (Msr1, also called SRA) is a pattern recognition receptor primarily expressed on myeloid cells, which plays an important role in the maintenance of immune homeostasis. Since MSR1 expression was upregulated in the livers of patients with fulminant hepatitis (FH), we investigated the functional mechanism of Msr1 in FH pathogenesis. METHODS: Msr1-deficient (Msr1-/-) mice and their wild-type (WT) littermates were infected with mouse hepatitis virus strain-A59 (MHV-A59) to induce FH, and the levels of tissue damage, serum alanine aminotransferase, inflammatory cytokines and complement component 5a (C5a) were measured and compared. Liver injury was studied after MHV infection with or without neutrophil depletion. RESULTS: Our results showed that Msr1-/- mice were resistant to MHV-induced hepatitis. Treatment with the C5a receptor antagonist (C5aRa) diminished the differences in inflammatory responses and liver injury between MHV-infected wild-type and Msr1-/- mice, suggesting that C5a-induced pro-inflammatory response plays a critical role in the Msr1-mediated regulation of FH pathogenesis. We demonstrated that Msr1 efficiently enhanced transforming growth factor-activated kinase-1 phosphorylation in neutrophils upon MHV-A59 stimulation, thereby promoting the activation of the extracellular signal-regulated kinase pathway and subsequent NETosis formation. Moreover, we provided evidence that blockage of Msr1 attenuated the liver damage caused by MHV-A59 infection. CONCLUSIONS: Msr1 promotes the pathogenesis of virus-induced FH by enhancing induction of neutrophil NETosis and subsequent complement activation. Targeting Msr1 may be employed as a new immunotherapeutic strategy for FH. LAY SUMMARY: Virus-induced fulminant hepatitis (FH) is a disease with a high mortality worldwide. Enhanced levels of macrophage scavenger receptor 1 (Msr1) in the liver of patients with FH and of murine experimental FH indicated Msr1 plays a role in the pathogenesis of FH. Herein, we demonstrate that mice deficient in Msr1 are resistant to FH induced by MHV-A59, and the Msr1 inhibitor fucoidan suppresses the progression of FH in mice. Our study suggests that use of drugs inhibiting MSR1 function could be beneficial to patients with FH.


Asunto(s)
Activación de Complemento , Hepatitis Viral Animal/etiología , Neutrófilos/fisiología , Receptores Depuradores de Clase A/fisiología , Animales , Complemento C5a/biosíntesis , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/terapia , Humanos , Quinasas Quinasa Quinasa PAM/fisiología , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina , Receptores Depuradores de Clase A/antagonistas & inhibidores
4.
Nat Commun ; 8(1): 1322, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-29109438

RESUMEN

Exacerbation of macrophage-mediated inflammation contributes to pathogenesis of various inflammatory diseases, but the immunometabolic programs underlying regulation of macrophage activation are unclear. Here we show that V-set immunoglobulin-domain-containing 4 (VSIG4), a B7 family-related protein that is expressed by resting macrophages, inhibits macrophage activation in response to lipopolysaccharide. Vsig4 -/- mice are susceptible to high-fat diet-caused obesity and murine hepatitis virus strain-3 (MHV-3)-induced fulminant hepatitis due to excessive macrophage-dependent inflammation. VSIG4 activates the PI3K/Akt-STAT3 pathway, leading to pyruvate dehydrogenase kinase-2 (PDK2) upregulation and subsequent phosphorylation of pyruvate dehydrogenase, which results in reduction in pyruvate/acetyl-CoA conversion, mitochondrial reactive oxygen species secretion, and macrophage inhibition. Conversely, interruption of Vsig4 or Pdk2 promotes inflammation. Forced expression of Vsig4 in mice ameliorates MHV-3-induced viral fulminant hepatitis. These data show that VSIG4 negatively regulates macrophage activation by reprogramming mitochondrial pyruvate metabolism.


Asunto(s)
Activación de Macrófagos/fisiología , Ácido Pirúvico/metabolismo , Receptores de Complemento/metabolismo , Animales , Infecciones por Coronavirus/etiología , Dieta Alta en Grasa/efectos adversos , Células HEK293 , Hepatitis Viral Animal/etiología , Humanos , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Virus de la Hepatitis Murina , Obesidad/etiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo , Receptores de Complemento/deficiencia , Receptores de Complemento/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células THP-1
5.
Biochim Biophys Acta ; 1842(9): 1464-71, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24768802

RESUMEN

UNLABELLED: Indoleamine 2,3-dioxygenase (IDO), an enzyme that is ubiquitously distributed in mammalian tissues and cells, converts tryptophan to kynurenine, and is also known as a key molecule that promotes apoptosis in lymphocytes and neurons. In this study, we established hepatitis B virus (HBV)-transgenic (Tg)/IDO-knockout (KO) mice and examined the influence of IDO in a murine fulminant hepatitis model induced by HBV-specific cytotoxic T lymphocytes (CTL). An increase of IDO expression in the livers of HBV-Tg/IDO-wild-type (WT) mice administered HBV-specific CTL was confirmed by real-time polymerase chain reaction, western blotting, and evaluating IDO activity. Plasma alanine aminotransferase (ALT) levels in HBV-Tg/IDO-KO mice after HBV-specific CTL injection significantly decreased compared with those in HBV-Tg/IDO-WT mice. An inhibitor of IDO, 1-methyl-d-tryptophan (1-MT), could also attenuated the observed liver injury induced by this HBV-specific CTL. The expression levels of cytokine and chemokine mRNAs in the livers of HBV-Tg/IDO-WT mice were higher than those in the livers of HBV-Tg/IDO-KO mice. The administration of kynurenine aggravated the liver injury in HBV-Tg/IDO-KO mice injected with HBV-specific CTL. Simultaneous injection of recombinant murine interferon (IFN-γ) and kynurenine also increased the ALT levels in HBV-Tg/IDO-KO mice. The liver injury induced by IFN-γ and kynurenine was improved in HBV-Tg/tumor necrosis factor-α-KO mice. CONCLUSION: Kynurenine and IFN-γ induced by the administration with HBV-specific CTL are cooperatively involved in the progression of liver injury in acute hepatitis model. Our results may lead to a new therapy for the acute liver injury caused by HBV infection.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Modelos Animales de Enfermedad , Virus de la Hepatitis B/patogenicidad , Hepatitis Viral Animal/etiología , Indolamina-Pirrol 2,3,-Dioxigenasa/fisiología , Quinurenina/toxicidad , Linfocitos T Citotóxicos/inmunología , Animales , Western Blotting , Proliferación Celular , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Hepatitis Viral Animal/enzimología , Hepatocitos/enzimología , Hepatocitos/inmunología , Hepatocitos/patología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Citotóxicos/trasplante
6.
J Virol ; 86(23): 12879-90, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22993151

RESUMEN

One common sign of human cytomegalovirus infection is altered liver function. Murine cytomegalovirus strain v70 induces a rapid and severe hepatitis in immunocompetent mice that requires the presence of T cells in order to develop. v70 exhibits approximately 10-fold-greater virulence than the commonly used strain K181, resulting in a more severe, sustained, and lethal hepatitis but not dramatically higher viral replication levels. Hepatitis and death are markedly delayed in immunodeficient SCID compared to immunocompetent BALB/c mice. Transfer of BALB/c splenocytes to SCID mice conferred rapid disease following infection, and depletion of either CD4 or CD8 T cells in BALB/c mice reduced virus-induced hepatitis. The frequency of CD8 T cells producing gamma interferon and tumor necrosis factor in response to viral antigen was higher in settings where more severe disease occurred. Thus, virus-specific effector CD8 T cells appear to contribute to lethal virus-induced hepatitis, contrasting their protective role during sublethal infection. This study reveals how protection and disease during cytomegalovirus infection depend on viral strain and dose, as well as the quality of the T cell response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Citomegalovirus/complicaciones , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/virología , Inmunidad Celular/inmunología , Muromegalovirus/patogenicidad , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Citometría de Flujo , Fluorescencia , Hepatitis Viral Animal/etiología , Técnicas Histológicas , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Estadísticas no Paramétricas , Factor de Necrosis Tumoral alfa/metabolismo
7.
Liver Int ; 29(2): 277-83, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18397228

RESUMEN

BACKGROUND/AIMS: Indoleamine-2,3-dioxygenase (IDO) is a tryptophan-catabolizing enzyme inducing suppression of T-cell function and immune tolerance. In hepatitis B virus (HBV) transgenic (Tg) mice, the adoptive transfer of HBV-specific cytotoxic T lymphocytes (CTL) causes a necroinflammatory liver disease that is histologically similar to acute viral hepatitis in man. The present study aimed to determine IDO expression in the liver and hepatocytes during an acute hepatitis model. METHODS: Serum l-kynurenine (l-Kyn) concentration in HBV Tg mice administered with HBV-specific CTL was measured over time, together with serum levels of alanine aminotransferase (ALT). Furthermore, we examined the expression of IDO in the total liver and isolated hepatocytes of HBV Tg mice after CTL injection using immunohistochemical analysis and reverse-transcription polymerase chain reaction (PCR). RESULTS: In HBV Tg mice, HBV-specific CTL induced, over the course of several days, a chronic increase in serum l-Kyn levels, which was associated with a sustained enhancement of liver IDO activity. In particular, IDO expression was enhanced in the liver parenchymal cells (hepatocytes) after HBV-specific CTL injection both in immunohistochemical analysis and in reverse-transcription PCR. Moreover, murine recombinant interferon-gamma (IFN-gamma) directly increased the IDO expression in primary hepatocytes in vitro. CONCLUSIONS: Cytotoxic T lymphocytes transduction results in the upregulation of IDO, which might downregulate T-cell responsiveness. Our findings provide evidence that hepatocyte itself expresses IDO and increases levels of l-Kyn in the blood in acute lethal hepatitis of mice. These data indicate that HBV infection facilitates the induction of IDO in response to proinflammatory cytokines, particularly IFN-gamma.


Asunto(s)
Virus de la Hepatitis B/inmunología , Hepatitis Viral Animal/enzimología , Hepatitis Viral Animal/etiología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/trasplante , Regulación hacia Arriba , Alanina Transaminasa/sangre , Animales , Hepatitis Viral Animal/inmunología , Hepatocitos/metabolismo , Inmunohistoquímica , Interferón gamma/metabolismo , Quinurenina/sangre , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
J Virol ; 78(18): 10156-65, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15331748

RESUMEN

CEACAM1a glycoproteins are members of the immunoglobulin (Ig) superfamily and the carcinoembryonic antigen family. Isoforms expressing either two or four alternatively spliced Ig-like domains in mice have been found in a number of epithelial, endothelial, or hematopoietic tissues. CEACAM1a functions as an intercellular adhesion molecule, an angiogenic factor, and a tumor cell growth inhibitor. Moreover, the mouse and human CEACAM1a proteins are targets of viral or bacterial pathogens, respectively, including the murine coronavirus mouse hepatitis virus (MHV), Haemophilus influenzae, Neisseria gonorrhoeae, and Neisseria meningitidis, as well as Moraxella catarrhalis in humans. We have shown that targeted disruption of the Ceacam1a (MHVR) gene resulting in a partial ablation of the protein in mice (p/p mice) led to reduced susceptibility to MHV-A59 infection of the modified mice in the BALB/c background. We have now engineered and produced a Ceacam1a-/- mouse that exhibits complete ablation of the CEACAM1a protein in every tissue where it is normally expressed. We report that 3-week-old Ceacam1a-/- mice in the C57BL/6 genetic background are fully resistant to MHV-A59 infection by both intranasal and intracerebral routes. Whereas virus-inoculated wild-type +/+ C57BL/6 mice showed profound liver damage and spinal cord demyelination under these conditions, Ceacam1a-/- mice displayed normal livers and spinal cords. Virus was recovered from liver and spinal cord tissues of +/+ mice but not of -/- mice. These results indicate that CEACAM1a is the sole receptor for MHV-A59 in both liver and brain and that its deletion from the mouse renders the mouse completely resistant to infection by this virus.


Asunto(s)
Glicoproteínas/deficiencia , Virus de la Hepatitis Murina/patogenicidad , Receptores Virales/deficiencia , Animales , Antígenos CD , Secuencia de Bases , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/fisiología , Moléculas de Adhesión Celular , Infecciones por Coronavirus/etiología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , ADN/genética , Marcación de Gen , Glicoproteínas/genética , Glicoproteínas/fisiología , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Virales/genética , Receptores Virales/fisiología
9.
J Virol ; 78(8): 3977-83, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15047813

RESUMEN

Viruses can spread by different mechanisms: via intracellular particles through cell junctions to neighboring cells or via secreted virions to adjacent or remote cells. The observation of clusters of hepadnavirus-infected cells both in vivo and in primary hepatocytes neither proves the first mechanism nor excludes the second. In order to test which mechanism, if not both, is used by hepatitis B viruses in order to spread, we used primary duck hepatocytes and duck hepatitis B virus (DHBV) as an infection model. If extracellular progeny virus alone determines spreading, neutralizing antisera or drugs blocking virus binding to hepatocytes should abolish secondary infection. In order to test this, we used DHBV envelope-specific neutralizing antisera, as well as suramin, a known inhibitor of infection. Both reagents strongly reduced hepatocellular attachment of viral particles and almost completely abolished primary infection, whereas an ongoing intracellular infection was not affected as long as no progeny virus was released. In contrast, incubation of infected primary hepatocytes with these reagents during release of progeny virus completely prevented secondary infection. Moreover, the combination of electron and immunofluorescence microscopy analyses revealed the residence of viral particles in cytoplasmic vesicles preferentially located near the basolateral membrane of infected hepatocytes. Taken together, these data strongly suggest that hepatitis B viruses mainly spread by secreted, extracellular progeny and point to polarized egress of viral particles into intercellular compartments, which restricts their diffusion and favors transmission of virus to adjacent cells.


Asunto(s)
Virus de la Hepatitis B del Pato/fisiología , Animales , Anticuerpos Antivirales/administración & dosificación , Células Cultivadas , Patos , Infecciones por Hepadnaviridae/etiología , Infecciones por Hepadnaviridae/virología , Virus de la Hepatitis B del Pato/inmunología , Virus de la Hepatitis B del Pato/patogenicidad , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/virología , Hepatocitos/efectos de los fármacos , Hepatocitos/virología , Técnicas In Vitro , Cuerpos de Inclusión Viral/ultraestructura , Microscopía Electrónica , Pruebas de Neutralización , Suramina/farmacología , Replicación Viral
10.
Comp Med ; 54(6): 681-9, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15679267

RESUMEN

Mouse hepatitis virus (MHV) is one of the most prevalent viruses infecting laboratory mice. Most natural infections are caused by enterotropic strains. Experiments were done to compare the pathogenesis of enterotropic strain MHV-Y in immunocompetent BALB/c and C57BL/6 mice with that in B and T cell-deficient mice. In situ hybridization was used to identify sites of virus replication, and reverse transcriptase-polymerase chain reaction analysis was used to detect viral RNA in feces and blood. MHV-Y caused acute subclinical infections restricted to the gastrointestinal tract in BALB/c and C57BL/6 mice. Viral RNA was detected in small intestine and associated lymphoid tissues of immunocompetent mice for 1 week and in cecum and colon for 2 weeks. Infected B cell-deficient mice developed chronic subclinical infection also restricted to the gastrointestinal tract. Viral RNA was detected in the small intestine, cecum, colon, and feces for 7 to 8 weeks. In contrast, infected T cell-deficient mice developed multisystemic lethal infection. During the first week, viral RNA was restricted to the gastrointestinal tract. However, by 2 weeks, mice developed peritonitis, and viral RNA was detected in mesentery and visceral peritoneum. Three to four weeks after virus inoculation, T cell-deficient mice became moribund and viral RNA was detected in multiple organ systems. These results suggest that B cells promote clearance of MHV-Y from intestinal mucosa and that T cells are required to prevent dissemination of MHV-Y from the gastrointestinal tract and associated lymphoid tissues.


Asunto(s)
Infecciones por Coronavirus/etiología , Infecciones por Coronavirus/inmunología , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/inmunología , Virus de la Hepatitis Murina/patogenicidad , Animales , Linfocitos B/inmunología , Femenino , Inmunocompetencia , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/inmunología , Hibridación in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/inmunología , Virus de la Hepatitis Murina/aislamiento & purificación , ARN Viral/genética , ARN Viral/aislamiento & purificación , Especificidad de la Especie , Linfocitos T/inmunología
11.
J Vet Med Sci ; 65(9): 1041-5, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-14532704

RESUMEN

Five 2 day-old colostrum-deprived piglets were inoculated with tissue homogenates from pigs with postweaning multisystemic wasting syndrome. One of the five piglets developed icterus and died 23 days post-inoculation. Histologic examination revealed acute hepatitis. Porcine circovirus type 2 (PCV-2) antigen and nucleic acid were detected in hepatocytes and phagocytic cells. Ultrastructurally, hepatocytes and phagocytic cells had large numbers of cytoplasmic inclusions, which were composed of electron-dense paracrystalline arrays of small non-enveloped viral particles approximately 17 nm in diameter. Apoptotic hepatocytes were confirmed by the TUNEL method and electron microscopic examination. These findings may indicate that hepatocellular necrosis is associated with replication of PCV-2. Apoptosis of hepatocytes also contributes to the pathogenesis of hepatic lesions in this case.


Asunto(s)
Hepatitis Viral Animal/etiología , Extractos de Tejidos/farmacología , Síndrome Debilitante/veterinaria , Enfermedad Aguda , Animales , Infecciones por Circoviridae/patología , Infecciones por Circoviridae/transmisión , Infecciones por Circoviridae/veterinaria , Hepatitis Viral Animal/patología , Hepatocitos/patología , Hepatocitos/ultraestructura , Hibridación in Situ , Porcinos , Enfermedades de los Porcinos/fisiopatología , Síndrome Debilitante/complicaciones , Síndrome Debilitante/virología , Destete
12.
Proc Natl Acad Sci U S A ; 100(17): 9962-7, 2003 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12907703

RESUMEN

Progress in understanding the pathogenesis of hepatitis C virus (HCV) has been slowed by the absence of tractable small animal models. Whereas GB virus B (GBV-B, an unclassified flavivirus) shares a phylogenetic relationship and several biologic attributes with HCV, including hepatotropism, it is not known to cause persistent infection, a hallmark of HCV. Here, we document persistent GBV-B infection in one of two healthy tamarins (Saguinus oedipus) inoculated intrahepatically with infectious synthetic RNA. High-titer viremia (108 to 109 genome equivalents per ml) and transiently elevated serum alanine transaminase activities were present from weeks 4 to 12 postinoculation in both animals. However, whereas GBV-B was eliminated from one animal by 20 weeks, the second animal remained viremic (103 to 107 genome equivalents per ml) for >2 years, with alanine transaminase levels becoming elevated again before spontaneous resolution of the infection. A liver biopsy taken late in the course of infection demonstrated hepatitis with periportal mononuclear infiltrates, hepatocellular microvesicular changes, cytoplasmic lipid droplets, and disordered mitochondrial ultrastructure, findings remarkably similar to chronic hepatitis C. GBV-B-infected hepatocytes contained numerous small vesicular membranous structures resembling those associated with expression of HCV nonstructural proteins, and sequencing of GBV-B RNA demonstrated a rate of molecular evolution comparable to that of HCV. We conclude that GBV-B is capable of establishing persistent infections in healthy tamarins, a feature that substantially enhances its value as a model for HCV. Mitochondrial structural changes and altered lipid metabolism leading to steatosis are conserved features of the pathogenesis of chronic hepatitis caused by these genetically distinct flaviviruses.


Asunto(s)
Infecciones por Flaviviridae/etiología , Virus GB-B/genética , Virus GB-B/patogenicidad , Hepatitis Viral Animal/etiología , ARN Viral/administración & dosificación , ARN Viral/genética , Saguinus/virología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Infecciones por Flaviviridae/patología , Infecciones por Flaviviridae/virología , Genoma Viral , Hepacivirus/genética , Hepacivirus/patogenicidad , Hepatitis C Crónica/etiología , Hepatitis Viral Animal/patología , Hepatitis Viral Animal/virología , Humanos , Hígado/patología , Datos de Secuencia Molecular , Mutación , Factores de Tiempo
13.
J Clin Invest ; 112(1): 58-66, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12840059

RESUMEN

Fibrin deposition and thrombosis within the microvasculature is now appreciated to play a pivotal role in the hepatocellular injury observed in experimental and human viral hepatitis. Importantly, the pathways by which fibrin generation is elicited in viral hepatitis may be mechanistically distinct from the classical pathways of coagulation induced by mechanical trauma or bacterial lipopolysaccharide (LPS). In the setting of murine hepatitis virus strain-3 (MHV-3) infection, a member of the Coronaviridae, activated endothelial cells and macrophages express distinct cell-surface procoagulants, including a novel prothrombinase, Fgl2/fibroleukin, which are important for both the initiation and localization of fibrin deposition. To assess the role of Fgl2/fibroleukin in murine viral hepatitis we generated a Fgl2/fibroleukin-deficient mouse. Peritoneal macrophages isolated from Fgl2/fibroleukin-/- mice did not generate a procoagulant response when infected with MHV-3. Fibrin deposition and liver necrosis were markedly reduced, and survival was increased in mice infected with MHV-3. To address the relevance of Fgl2/fibroleukin in human chronic viral hepatitis we studied patients with minimal and marked chronic hepatitis B. We detected robust expression of Fgl2/fibroleukin mRNA transcripts and protein in liver tissue isolated from patients with marked chronic hepatitis B. Fibrin deposition was strongly associated with Fgl2/fibroleukin expression. Collectively, these data indicate a critical role for Fgl2/fibroleukin in the pathophysiology of experimental and human viral hepatitis.


Asunto(s)
Fibrinógeno/fisiología , Hepatitis Viral Animal/etiología , Hepatitis Viral Humana/etiología , Trombosis/etiología , Adulto , Animales , Susceptibilidad a Enfermedades , Femenino , Fibrinógeno/genética , Hemorragia/etiología , Hepatitis Crónica/complicaciones , Hepatitis Crónica/metabolismo , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Humana/metabolismo , Humanos , Lipopolisacáridos/toxicidad , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , ARN Mensajero/análisis
14.
FASEB J ; 17(1): 94-6, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12475902

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is able to kill a broad spectrum of tumor cells but appears to be nontoxic to most normal cells. Because there are conflicting data about the hepatotoxicity of TRAIL, we investigated the physiological function of TRAIL and its receptors in the liver. Hepatocytes are sensitive for FasL- and TRAIL-mediated apoptosis in vitro, but TRAIL induces no apoptosis in healthy livers in vivo. Using mouse models of adenoviral hepatitis and livers of patients with hepatitis infection, we could demonstrate that apoptosis in virally infected hepatocytes is mediated by TRAIL receptor DR5 and TRAIL. In contrast to FasL, TRAIL-mediated apoptosis of hepatocytes in vivo is triggered through viral infection. The TRAIL receptor/ligand system enables the organisms to specifically kill virus-infected hepatocytes, whereas normal uninfected hepatocytes in vivo are resistant to TRAIL-mediated apoptosis. Overexpression of TRAIL in the liver after viral infection is not dependent on lymphocytes, natural killer, or Kupffer cells, which indicates that the TRAIL receptor/ligand system is a paracrine mechanism of hepatocytes against virally infected cells. Our results suggest that TRAIL might be used not only for cancer therapy but also for therapy of patients with viral hepatitis to selectively eliminate infected hepatocytes and limit viral replication.


Asunto(s)
Apoptosis , Hepatitis Viral Humana/etiología , Glicoproteínas de Membrana/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología , Factor de Necrosis Tumoral alfa/fisiología , Adenoviridae/patogenicidad , Infecciones por Adenoviridae/patología , Infecciones por Adenoviridae/virología , Animales , Proteínas Reguladoras de la Apoptosis , Células Cultivadas , Proteína Ligando Fas , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/virología , Hepatitis Viral Humana/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Fallo Hepático Agudo/etiología , Fallo Hepático Agudo/metabolismo , Glicoproteínas de Membrana/farmacología , Glicoproteínas de Membrana/toxicidad , Ratones , Modelos Biológicos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/biosíntesis , Receptores del Factor de Necrosis Tumoral/genética , Ligando Inductor de Apoptosis Relacionado con TNF , Activación Transcripcional , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba
15.
Front Biosci ; 7: d1899-914, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12161342

RESUMEN

Chemokines may be involved in the tissue response to injury regulating the influx of leukocytes, and modulating a number of other critical biologic actions, including angiogenesis, neoplastic growth, myo-fibroblast activation, and the response to viral infections. In the liver, up-regulated expression of different members of the chemokine system may be induced by almost all types of injury, and there is often a clear relation between the chemokine pattern activated by different types of injury and the predominant subclasses of leukocytes which infiltrate the liver. Neutralization of specific chemokines by passive immunization or the use of animals deficient in specific chemokines or chemokine receptors has indicated a causal relation between up-regulation of chemokines and leukocyte infiltration. Inflammation is part of the liver wound healing response, that in chronic conditions leads to the development of fibrosis and cirrhosis. Hepatic stellate cells, which play a leading role in the development of fibrosis following their transition to myofibroblasts, express different chemokines. Chemokine expression by stellate cells is regulated by soluble mediators, in particular pro-inflammatory cytokines, as well as growth factors, proteases, and products of oxidative stress. In addition, stellate cells also respond to chemokines with biologic actions relevant for tissue repair, such as cell migration or induction of other chemokines. These data indicate that chemokines in the liver may modulate the progression of liver fibrosis through actions on hepatic stellate cells.


Asunto(s)
Quimiocinas/fisiología , Hepatitis Animal/etiología , Cirrosis Hepática/etiología , Animales , Progresión de la Enfermedad , Rechazo de Injerto/etiología , Hepatitis Alcohólica/etiología , Hepatitis Viral Animal/etiología , Hígado/citología , Hígado/metabolismo , Neoplasias Hepáticas/etiología , Trasplante de Hígado , Modelos Biológicos , Daño por Reperfusión/etiología
16.
Prostate Cancer Prostatic Dis ; 5(4): 316-25, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12627218

RESUMEN

Adenoviral vector delivery of the Herpes simplex virus thymidine kinase (HSV-tk) gene in combination with the prodrug ganciclovir (GCV) has been tested in phase I clinical trials for prostate cancer and found to exhibit a satisfactory toxicity profile. We have developed additional adenoviral vectors with differing promoters to optimize the expression profile and in the present study evaluate the potential systemic toxicity of these vectors. Four recombinant adenoviral vectors that express the HSV-tk gene were generated using three different promoters: CMV (leftward orientation); RSV (both rightward and leftward orientation); and the mouse caveolin-1 (cav-1) promoter (leftward orientation). Efficacy was determined in vitro by cytotoxicity assays in a mouse prostate cancer cell line, RM-9, and in vivo by treating orthotopic tumors. Potential toxicity was evaluated from liver histology and apoptotic cell counts and enzyme levels in the serum following intravenous adenoviral vector injection. Although there were differences in HSV-tk expression at the protein level among the four vectors there were no significant differences in in-vitro cytotoxicity studies with GCV or in vivo in tumor growth suppression of an orthotopic mouse prostate cancer model in GCV treated mice. Intravenous delivery of high doses of all adenoviral vectors lead to abnormalities in liver function as measured by specific serum markers and histological evaluation of liver tissue and increased levels of apoptosis in the liver. These abnormalities were most prevalent with the vector containing the CMV promoter and the rightward oriented RSV promoter. They were least prevalent in the vector regulated by the cav-1 promoter. Upregulation of specific chemokines, MIP-2 and MIP-1beta was correlated with apoptotic counts. Our results demonstrate that comprehensive toxicological analysis of adenoviral vectors provides internally consistent information that can differentiate vectors with comparable efficacy based on toxicity. In these studies vectors with the cav-1 promoter-driven and leftward RSV-driven HSV-tk gene demonstrated minimal toxicities with cytotoxic effectiveness comparable to more toxic vectors. Our studies further suggest that promoter selection can influence the toxic effects of an adenoviral gene therapy vector.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/genética , Virus del Sarcoma Aviar/genética , Caveolinas/genética , Quimiocinas/biosíntesis , Citomegalovirus/genética , Virus Defectuosos/genética , Genes Virales , Terapia Genética , Vectores Genéticos/toxicidad , Hepatitis Viral Animal/etiología , Regiones Promotoras Genéticas , Neoplasias de la Próstata/terapia , Simplexvirus/genética , Timidina Quinasa/genética , Proteínas Virales/genética , Animales , Apoptosis , Caveolina 1 , Quimiocina CCL4 , Quimiocina CCL5/biosíntesis , Quimiocina CCL5/genética , Quimiocina CXCL10 , Quimiocina CXCL2 , Quimiocinas/genética , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Expresión Génica , Regulación Viral de la Expresión Génica , Genes Sintéticos , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Inyecciones Intravenosas , Pruebas de Función Hepática , Proteínas Inflamatorias de Macrófagos/biosíntesis , Proteínas Inflamatorias de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Monocinas/biosíntesis , Monocinas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simplexvirus/enzimología , Timidina Quinasa/antagonistas & inhibidores , Células Tumorales Cultivadas/patología , Células Tumorales Cultivadas/virología , Proteínas Virales/antagonistas & inhibidores
17.
J Gen Virol ; 82(Pt 10): 2437-2448, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11562537

RESUMEN

The strong similarity between GB virus B (GBV-B) and hepatitis C virus (HCV) makes tamarins infected by GBV-B an acceptable surrogate animal model for HCV infection. Even more attractive, for drug discovery purposes, is the idea of constructing chimeric viruses by inserting HCV genes of interest into a GBV-B genome frame. To accomplish this, infectious cDNA clones of both viruses must be available. The characterization of several HCV molecular clones capable of infecting chimpanzees has been published, whereas only one infectious GBV-B clone inducing hepatitis in tamarins has been reported so far. Here we describe the infection of tamarins by intrahepatic injection of RNA transcribed from a genomic GBV-B clone (FL-3) and transmission of the disease from infected to naive tamarins via serum inoculation. The disease resulting from both direct and secondary infection was characterized for viral RNA titre and hepatitis parameters as well as for viral RNA distribution in the hepatic tissue. Host humoral immune response to GBV-B antigens was also monitored. The progression of the disease was compared to that induced by intravenous injection of different amounts of the non-recombinant virus.


Asunto(s)
Flaviviridae/genética , Hepatitis Viral Animal/etiología , Virión/genética , Animales , Secuencia de Bases , Genoma Viral , Anticuerpos Antihepatitis/biosíntesis , Leucocitos Mononucleares/virología , Hígado/virología , Datos de Secuencia Molecular , ARN Viral/análisis , Saguinus , Transcripción Genética
18.
Avian Dis ; 45(1): 215-22, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11336070

RESUMEN

The hypothesis that fowl adenovirus (FAV) and chicken anemia virus (CAV), transmitted vertically and simultaneously, induce the inclusion body hepatitis (IBH)/hydropericardium (HP) syndrome in progeny chickens was tested. Thus, 35-wk-old light brown layer breeders, showing absence of antibodies against FAV and variable titers against CAV, were intramuscularly singly infected with the FAV serotype 4 isolate 341 or dually infected with CAV (isolate 10343) and FAV. All hens (groups A [FAV alone], B [FAV + CAV], and C [noninfected]) were clinically healthy throughout the experimental period. Both infectious viruses FAV and CAV were isolated from progenies obtained as early as 5 days after infection of their breeders. Hematocrit, serum proteins, and aspartate-aminotransferase values showed a few statistical differences between the progeny groups. Most of these differences were detected in the progeny chickens of group B. However, almost all values met reference values for the species. The pathologic findings showed that progeny chickens obtained from both singly and dually infected breeders developed macroscopic and histopathologic changes of IBH/HP. The pathologic findings shown by progeny chickens of group A (FAV) were not expected because neither synergism nor prior immunodepression by CAV was concurrent. Chickens of group B (CAV + FAV) also developed IBH/HP. Although not many differences in the evaluated parameters between groups A and B were statistically significant, most pathologic findings of group B indicated a more severe manifestation of the disease. However, because FAV alone did reproduce the syndrome, the results shown by group B would not allow a definitive confirmation of the hypothesis that the association of FAV and CAV is necessary for the successful induction of the IBH/HP syndrome in chickens when transmitted vertically.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Infecciones por Circoviridae/veterinaria , Hepatitis Viral Animal/etiología , Cuerpos de Inclusión Viral/virología , Transmisión Vertical de Enfermedad Infecciosa/veterinaria , Enfermedades de las Aves de Corral/etiología , Infecciones por Adenoviridae/complicaciones , Animales , Aviadenovirus , Virus de la Anemia del Pollo , Pollos , Infecciones por Circoviridae/complicaciones , Hepatitis Viral Animal/patología , Cuerpos de Inclusión Viral/patología , Riñón/patología , Hígado/patología , Pericardio/patología , Pericardio/virología
19.
J Immunol ; 165(5): 2415-22, 2000 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10946266

RESUMEN

To study peripheral tolerance of CD8 T cells to a classically MHC-restricted peptide Ag expressed in hepatocytes, ALB1 transgenic (tg) mice expressing the CTL epitope GP33 of the lymphocytic choriomeningitis virus glycoprotein under control of the mouse albumin promoter were generated. ALB1 mice exclusively expressed the GP33 transgene in the liver and, at a 100- to 1000-fold lower level, in the thymus. TCR-tg mice specific for the GP33 epitope were used to directly follow GP33-specific T cells in vivo. These experiments revealed that 1) thymic expression of the GP33 transgene led to incomplete central deletion of TCR-tg cells; and 2) peripheral TCR-tg cells in ALB1 mice ignored the GP33 transgene expressed in hepatocytes. Ignorance of adoptively transferred TCR-tg cells in ALB1 mice was broken by infection with lymphocytic choriomeningitis virus, leading to induction of hepatitis in ALB1, but not in control, mice. Taken together, we have established a novel model of virus-induced CD8 T cell-mediated autoimmune hepatitis in mice and demonstrate that naive CD8 T cells may ignore Ags expressed in the liver.


Asunto(s)
Antígenos Virales/inmunología , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/inmunología , Tolerancia Inmunológica , Hígado/inmunología , Hígado/virología , Linfocitos T/inmunología , Proteínas Virales , Traslado Adoptivo , Animales , Antígenos Virales/biosíntesis , Antígenos Virales/genética , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Hepatitis Viral Animal/genética , Tolerancia Inmunológica/genética , Hígado/metabolismo , Depleción Linfocítica , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Transgénicos , Fragmentos de Péptidos/biosíntesis , Fragmentos de Péptidos/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/metabolismo , Timo/inmunología , Timo/metabolismo , Transgenes/inmunología
20.
J Clin Invest ; 105(6): 813-21, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10727450

RESUMEN

We assessed the effect of modified antigen presenting cells (APCs) expressing high levels of Fas ligand (APC-FasL) on post-viral chronic inflammatory disease. FasL-deficient B6-gld/gld mice infected with murine cytomegalovirus (MCMV) cleared the virus from their lungs, kidneys, and livers within 2 weeks of infection. However, inflammation persisted in these organs for more than 8 weeks, with a chronically increased T-cell response to MCMV-infected APCs and production of autoantibodies. Administration of APC-AdFasL at 4 weeks suppressed this inflammation and diminished the T-cell response and autoantibody production. APC-AdFasL that had been transfected with ultraviolet-irradiated MCMV were more effective than uninfected APC-AdFasL in ameliorating the chronic inflammation. APC-AdFasL migrated preferentially to the spleen, where they triggered apoptosis of lymphocytes in the marginal zone of the spleen. These results confirm that Fas-mediated apoptosis is not required for clearance of virus, but is required for down-modulation of the virally induced chronic inflammatory response. This organwide effect of APC-AdFasL appears to be mediated by elimination of activated T lymphocytes in the spleen before their emigration to the target organs.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Apoptosis , Infecciones por Citomegalovirus/inmunología , Glicoproteínas de Membrana/fisiología , Adenoviridae/genética , Animales , Células Presentadoras de Antígenos/trasplante , Autoanticuerpos/biosíntesis , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/terapia , Proteína Ligando Fas , Femenino , Vectores Genéticos/genética , Hepatitis Viral Animal/etiología , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/patología , Inflamación , Riñón/patología , Hígado/patología , Pulmón/patología , Enfermedades Pulmonares Intersticiales/etiología , Enfermedades Pulmonares Intersticiales/inmunología , Enfermedades Pulmonares Intersticiales/patología , Macrófagos/inmunología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Ratones Mutantes , Nefritis/etiología , Nefritis/inmunología , Nefritis/patología , Organismos Libres de Patógenos Específicos , Bazo/inmunología , Bazo/patología , Subgrupos de Linfocitos T/inmunología , Transfección , Receptor fas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...