Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Más filtros













Intervalo de año de publicación
1.
Vet Res ; 55(1): 63, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760810

RESUMEN

The maintenance of viral protein homeostasis depends on the interaction between host cell proteins and viral proteins. As a molecular chaperone, heat shock protein 70 (HSP70) has been shown to play an important role in viral infection. Our results showed that HSP70 can affect translation, replication, assembly, and release during the life cycle of duck hepatitis A virus type 1 (DHAV-1). We demonstrated that HSP70 can regulate viral translation by interacting with the DHAV-1 internal ribosome entry site (IRES). In addition, HSP70 interacts with the viral capsid proteins VP1 and VP3 and promotes their stability by inhibiting proteasomal degradation, thereby facilitating the assembly of DHAV-1 virions. This study demonstrates the specific role of HSP70 in regulating DHAV-1 replication, which are helpful for understanding the pathogenesis of DHAV-1 infection and provide additional information about the role of HSP70 in infection by different kinds of picornaviruses, as well as the interaction between picornaviruses and host cells.


Asunto(s)
Proteínas HSP70 de Choque Térmico , Virus de la Hepatitis del Pato , Sitios Internos de Entrada al Ribosoma , Replicación Viral , Virus de la Hepatitis del Pato/fisiología , Virus de la Hepatitis del Pato/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Animales , Proteínas Estructurales Virales/metabolismo , Proteínas Estructurales Virales/genética , Patos , Enfermedades de las Aves de Corral/virología , Infecciones por Picornaviridae/veterinaria , Infecciones por Picornaviridae/virología , Infecciones por Picornaviridae/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/genética , Hepatitis Viral Animal/virología , Hepatitis Viral Animal/metabolismo , Biosíntesis de Proteínas
2.
Cell Mol Gastroenterol Hepatol ; 14(5): 1077-1101, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35926777

RESUMEN

BACKGROUND & AIMS: Fulminant viral hepatitis (FVH) is a life-threatening disease, but its pathogenesis is not fully understood. Neutrophil extracellular traps (NETs) were an unrecognized link between inflammation and coagulation, which are 2 main features of FVH. Here, we investigated the role and mechanism of NETs in the pathogenesis of FVH. METHODS: A mouse model of FVH was established by murine hepatitis virus strain-3 infection. Liver leukocytes of infected or uninfected mice were used for single-cell RNA sequencing and whole-transcriptome sequencing. NETs depletion was achieved using DNase 1. Acetaminophen was used to establish a mouse model of non-virus-caused acute liver failure. Clinically, NETs-related markers in liver, plasma, and peripheral neutrophils were assessed in patients with hepatitis B virus (HBV)-related acute liver injury. RESULTS: Increased hepatic NETs formation was observed in murine hepatitis virus strain-3-infected mice, but not in acetaminophen-treated mice. NETs depletion improved the liver damage and survival rate in FVH by inhibiting hepatic fibrin deposition and inflammation. An adoptive transfer experiment showed that neutrophil-specific fibrinogen-like protein 2 (FGL2) promoted NETs formation. FGL2 was found to directly interact with mucolipin 3, which regulated calcium influx and initiated autophagy, leading to NETs formation. Clinically, increased plasma NETs level was associated with coagulation dysfunction in patients with HBV acute liver injury. Colocalization of FGL2, NETs, and fibrin in liver was observed in these patients. CONCLUSIONS: NETs aggravated liver injury in FVH by promoting fibrin deposition and inflammation. NETs formation was regulated by the FGL2-mucolipin 3-autophagy axis. Targeting NETs may provide a new strategy for the treatment of FVH.


Asunto(s)
Trampas Extracelulares , Hepatitis Viral Animal , Hepatitis Viral Humana , Virus de la Hepatitis Murina , Ratones , Animales , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/patología , Ratones Endogámicos BALB C , Acetaminofén/efectos adversos , Calcio/metabolismo , Virus de la Hepatitis Murina/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Hepatitis Viral Humana/complicaciones , Modelos Animales de Enfermedad , Inflamación , Autofagia , Fibrina/metabolismo , Desoxirribonucleasas/metabolismo
3.
Microbiol Spectr ; 10(1): e0226521, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35138149

RESUMEN

Avian hepatitis E virus (HEV) causes liver diseases and multiple extrahepatic disorders in chickens. However, the mechanisms involved in avian HEV entry remain elusive. Herein, we identified the RAS-related protein 1b (Rap1b) as a potential HEV-ORF2 protein interacting candidate. Experimental infection of chickens and cells with an avian HEV isolate from China (CaHEV) led to upregulated expression and activation of Rap1b both in vivo and in vitro. By using CaHEV capsid as mimic of virion to treat cell in vitro, it appears that the interaction between the viral capsid and Rap1b promoted cell membrane recruitment of the downstream effector Rap1-interacting molecule (RIAM). In turn, RIAM further enhanced Talin-1 membrane recruitment and retention, which led to the activation of integrin α5/ß1, as well as integrin-associated membrane protein kinases, including focal adhesion kinase (FAK). Meanwhile, FAK activation triggered activation of downstream signaling molecules, such as Ras-related C3 botulinum toxin substrate 1 RAC1 cell division cycle 42 (CDC42), p21-activated kinase 1 (PAK1), and LIM domain kinase 1 (LIMK1). Finally, F-actin rearrangement induced by Cofilin led to the formation of lamellipodia, filopodia, and stress fibers, contributes to plasma membrane remodeling, and might enhance CaHEV virion internalization. In conclusion, our data suggested that Rap1b activation was triggered during CaHEV infection and appeared to require interaction between CaHEV-ORF2 and Rap1b, thereby further inducing membrane recruitment of Talin-1. Membrane-bound Talin-1 then activates key Integrin-FAK-Cofilin cascades involved in modulation of actin kinetics, and finally leads to F-actin rearrangement and membrane remodeling to potentially facilitate internalization of CaHEV virions into permissive cells. IMPORTANCE Rap1b is a multifunctional protein that is responsible for cell adhesion, growth, and differentiation. The inactive form of Rap1b is phosphorylated and distributed in the cytoplasm, while active Rap1b is prenylated and loaded with GTP to the cell membrane. In this study, the activation of Rap1b was induced during the early stage of avian HEV infection under the regulation of PKA and SmgGDS. Continuously activated Rap1b recruited its effector RIAM to the membrane, thereby inducing the membrane recruitment of Talin-1 that led to the activation of membrane α5/ß1 integrins. The triggering of the signaling pathway-associated Integrin α5/ß1-FAK-CDC42&RAC1-PAK1-LIMK1-Cofilin culminated in F-actin polymerization and membrane remodeling that might promote avian HEV virion internalization. These findings suggested a novel mechanism that is potentially utilized by avian HEV to invade susceptible cells.


Asunto(s)
Citoesqueleto/metabolismo , Hepatitis Viral Animal/metabolismo , Hepevirus/patogenicidad , Enfermedades de las Aves de Corral/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Proteínas de Unión al GTP rap/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Pollos , Citoesqueleto/genética , Citoesqueleto/virología , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/virología , Hepevirus/genética , Interacciones Huésped-Patógeno , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/virología , Unión Proteica , Proteínas Virales/genética , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/genética , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rap/genética
4.
Arch Virol ; 166(11): 3105-3116, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34482448

RESUMEN

Several outbreaks of duck hepatitis A virus type 1 (DHAV-1), which were characterized by yellow coloration and hemorrhage in pancreatic tissues, have occurred in China. The causative agent is called pancreatitis-associated DHAV-1. The mechanisms involved in pancreatitis-associated DHAV-1 infection are still unclear. Transcriptome analysis of duck pancreas infected with classical-type DHAV-1 and pancreatitis-associated DHAV-1 was carried out. Deep sequencing with Illumina-Solexa resulted in a total of 53.9 Gb of clean data from the cDNA library of the pancreas, and a total of 29,597 unigenes with an average length of 993.43 bp were generated by de novo sequence assembly. The expression levels of D-3-phosphoglycerate dehydrogenase, phosphoserine aminotransferase, and phosphoserine phosphatase, which are involved in glycine, serine, and threonine metabolism pathways, were significantly downregulated in ducks infected with pancreatitis-associated DHAV-1 compared with those infected with classical-type DHAV-1. These findings provide information regarding differences in expression levels of metabolism-associated genes between ducks infected with pancreatitis-associated DHAV-1 and those infected with classical-type DHAV-1, indicating that intensive metabolism disorders may contribute to the different phenotypes of DHAV-1-infection.


Asunto(s)
Virus de la Hepatitis del Pato/patogenicidad , Hepatitis Viral Animal/virología , Interacciones Huésped-Patógeno/genética , Infecciones por Picornaviridae/veterinaria , Enfermedades de las Aves de Corral/virología , Aminoácidos/genética , Aminoácidos/metabolismo , Animales , Patos/virología , Expresión Génica , Hepatitis Viral Animal/genética , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/patología , Páncreas/citología , Páncreas/patología , Páncreas/virología , Pancreatitis/patología , Pancreatitis/virología , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/patología , Infecciones por Picornaviridae/virología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN
5.
PLoS Pathog ; 16(10): e1008973, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33045014

RESUMEN

The liver is a central regulator of metabolic homeostasis and serum metabolite levels. Hepatocytes are the functional units of the liver parenchyma and not only responsible for turnover of biomolecules but also act as central immune signaling platforms. Hepatotropic viruses infect liver tissue, resulting in inflammatory responses, tissue damage and hepatitis. Combining well-established in vitro and in vivo model systems with transcriptomic analyses, we show that type I interferon signaling initiates a robust antiviral immune response in hepatocytes. Strikingly, we also identify IFN-I as both, sufficient and necessary, to induce wide-spread metabolic reprogramming in hepatocytes. IFN-I specifically rewired tryptophan metabolism and induced hepatic tryptophan oxidation to kynurenine via Tdo2, correlating with altered concentrations of serum metabolites upon viral infection. Infected Tdo2-deficient animals displayed elevated serum levels of tryptophan and, unexpectedly, also vast increases in the downstream immune-suppressive metabolite kynurenine. Thus, Tdo2-deficiency did not result in altered serum homeostasis of the tryptophan to kynurenine ratio during infection, which seemed to be independent of hepatocyte-intrinsic compensation via the IDO-axis. These data highlight that inflammation-induced reprogramming of systemic tryptophan metabolism is tightly regulated in viral hepatitis.


Asunto(s)
Antivirales/metabolismo , Hepatitis Viral Animal/inmunología , Hepatocitos/inmunología , Inflamación/inmunología , Quinurenina/metabolismo , Receptor de Interferón alfa y beta/fisiología , Triptófano/metabolismo , Animales , Femenino , Virus de Hepatitis/aislamiento & purificación , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/virología , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Inmunidad Innata/inmunología , Inflamación/metabolismo , Inflamación/patología , Inflamación/virología , Factor 7 Regulador del Interferón/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT1/fisiología , Triptófano Oxigenasa/fisiología
6.
J Virol ; 93(20)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31375571

RESUMEN

Mouse hepatitis virus (MHV) uses its N-terminal domain (NTD) of the viral spike (S) protein to bind the host receptor mouse carcinoembryonic antigen-related cell adhesion molecule 1a (mCEACAM1a) and mediate virus entry. Our previous crystal structure study of the MHV NTD/mCEACAM1a complex (G. Peng, D. Sun, K. R. Rajashankar, Z. Qian, et al., Proc Natl Acad Sci U S A 108:10696-10701, 2011, https://doi.org/10.1073/pnas.1104306108) reveals that there are 14 residues in the NTD interacting with the receptor. However, their contribution to receptor binding and virus entry has not been fully investigated. Here we analyzed 13 out of 14 contact residues by mutagenesis and identified I22 as being essential for receptor binding and virus entry. Unexpectedly, we found that G29 was critical for the conformational changes of the S protein triggered by either receptor binding or high pH. Replacement of G29 with A, D, F, K, M, and T, to different extents, caused spontaneous dissociation of S1 from the S protein, resulting in an enhancement of high-pH-triggered receptor-independent syncytium (RIS) formation in HEK293T cells, compared to the wild type (WT). In contrast, replacement of G29 with P, a turn-prone residue with a strict conformation, hindered virus entry and conformational changes of the S protein triggered by either receptor binding or pH 8.0, suggesting that the structural turn around G29 and its flexibility are critical. Finally, stabilization of the NTD by G29P had almost no effect on pH-independent RIS induced by the Y320A mutation in the C-terminal domain (CTD) of the S1 subunit, indicating that there might be an absence of cross talk between the NTD and CTD during conformational changes of the S protein. Our study will aid in better understanding the mechanism of how conformational changes of the S protein are triggered.IMPORTANCE Binding of the MHV S protein to the receptor mCEACAM1a triggers conformational changes of S proteins, leading to the formation of a six-helix bundle and viral and cellular membrane fusion. However, the mechanism by which the conformational change of the S protein is initiated after receptor binding has not been determined. In this study, we showed that while replacement of G29, a residue at the edge of the receptor binding interface and the center of the structural turn after the ß1-sheet of the S protein, with D or T triggered spontaneous conformational changes of the S protein and pH-independent RIS, the G29P mutation significantly impeded the conformational changes of S proteins triggered by either receptor binding or pH 8.0. We reason that this structural turn might be critical for conformational changes of the S protein and that altering this structural turn could initiate conformational changes of the S protein, leading to membrane fusion.


Asunto(s)
Glicina , Interacciones Huésped-Patógeno , Concentración de Iones de Hidrógeno , Virus de la Hepatitis Murina/fisiología , Receptores Virales/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Secuencia de Aminoácidos , Animales , Glicina/química , Glicina/genética , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/virología , Ratones , Modelos Moleculares , Unión Proteica , Conformación Proteica , Receptores Virales/química , Glicoproteína de la Espiga del Coronavirus/genética , Relación Estructura-Actividad
7.
Cell Death Dis ; 10(1): 12, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622241

RESUMEN

The protein kinase RIPK1 plays a crucial role at the crossroad of stress-induced signaling pathways that affects cell's decision to live or die. The present study aimed to define the role of RIPK1 in hepatocytes during fulminant viral hepatitis, a worldwide syndrome mainly observed in hepatitis B virus (HBV) infected patients. Mice deficient for RIPK1, specifically in liver parenchymal cells (Ripk1LPC-KO) and their wild-type littermates (Ripk1fl/fl), were challenged by either the murine hepatitis virus type 3 (MHV3) or poly I:C, a synthetic analog of double-stranded RNA mimicking viral pathogen-associated molecular pattern. Ripk1LPC-KO mice developed more severe symptoms at early stage of the MHV3-induced fulminant hepatitis. Similarly, administration of poly I:C only triggered increase of systemic transaminases in Ripk1LPC-KO mice, reflecting liver damage through induced apoptosis as illustrated by cleaved-caspase 3 labeling of liver tissue sections. Neutralization of TNF-α or prior depletion of macrophages were able to prevent the appearance of apoptosis of hepatocytes in poly I:C-challenged Ripk1LPC-KO mice. Moreover, poly I:C never induced direct hepatocyte death in primary culture whatever the murine genotype, while it always stimulated an anti-viral response. Our investigations demonstrated that RIPK1 protects hepatocytes from TNF-α secreted from macrophages during viral induced fulminant hepatitis. These data emphasize the potential worsening risks of an HBV infection in people with polymorphism or homozygous amorphic mutations already described for the RIPK1 gene.


Asunto(s)
Hepatitis Viral Animal/metabolismo , Hepatocitos/metabolismo , Hepatopatías/metabolismo , Necrosis Hepática Masiva/metabolismo , Virus de la Hepatitis Murina , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Células Cultivadas , Ácido Clodrónico/farmacología , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Hepatocitos/efectos de los fármacos , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hepatopatías/virología , Necrosis Hepática Masiva/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poli I-C/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
8.
PLoS Pathog ; 14(4): e1006989, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29652922

RESUMEN

The OAS/RNase L pathway is one of the best-characterized effector pathways of the IFN antiviral response. It inhibits the replication of many viruses and ultimately promotes apoptosis of infected cells, contributing to the control of virus spread. However, viruses have evolved a range of escape strategies that act against different steps in the pathway. Here we unraveled a novel escape strategy involving Theiler's murine encephalomyelitis virus (TMEV) L* protein. Previously we found that L* was the first viral protein binding directly RNase L. Our current data show that L* binds the ankyrin repeats R1 and R2 of RNase L and inhibits 2'-5' oligoadenylates (2-5A) binding to RNase L. Thereby, L* prevents dimerization and oligomerization of RNase L in response to 2-5A. Using chimeric mouse hepatitis virus (MHV) expressing TMEV L*, we showed that L* efficiently inhibits RNase L in vivo. Interestingly, those data show that L* can functionally substitute for the MHV-encoded phosphodiesterase ns2, which acts upstream of L* in the OAS/RNase L pathway, by degrading 2-5A.


Asunto(s)
2',5'-Oligoadenilato Sintetasa/metabolismo , Nucleótidos de Adenina/metabolismo , Endorribonucleasas/antagonistas & inhibidores , Virus de la Hepatitis Murina/fisiología , Oligorribonucleótidos/metabolismo , Theilovirus/metabolismo , Proteínas Virales/metabolismo , Animales , Antivirales/metabolismo , Endorribonucleasas/fisiología , Células HeLa , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/virología , Interacciones Huésped-Patógeno , Humanos , Ratones
9.
Avian Dis ; 62(1): 57-64, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29620455

RESUMEN

The aim of the present study was to further unravel the pathophysiologic mechanisms of inclusion body hepatitis (IBH). In a first trial, the susceptibility of specific-pathogen-free (SPF) broilers to fowl aviadenovirus (FAdV) infections was investigated. Regardless of viral dose, route of infection, and susceptibility to disease on day 1, the 3-week-old SPF broilers showed resistance to IBH, with no mortality being recorded throughout the experiment. In a second trial, SPF broilers were orally infected at 3 weeks of age with a FAdV-E strain, and their digestive and metabolic processes were monitored. The birds' performance decreased from 7 days postinfection (dpi) onward, and hepato- and pancreatomegaly were found at necropsy at 4, 7, and 10 dpi and at 7 dpi, respectively. Clinical chemistry revealed transient hyperlipasemia at 4 dpi and hyperglycemia from 4 dpi onwards, with 25% of infected birds showing glycemia levels suggestive of diabetes mellitus. Histopathology findings included typical adenoviral hepatitis in the liver, while in the pancreas, inflammation characterized by multifocal infiltrations of lymphocytes, together with shrinkage of acinar cells, loss of acinar arrangement, and hyperplasia of islet cells, was noticed. Additionally, the pancreatic tissue had tendentiously lower levels of enzyme activity, and in the ileum, the digestibility of fat was significantly impaired. Hence, our data reinforce the concept of age-related resistance to experimentally induced IBH. Additionally, we demonstrated that FAdV-induced pancreatitis in broilers interferes with the digestive process and evolves into a dysmetabolic condition that resembles diabetes, affecting the health and zootechnical performance of birds, and therefore providing an important component of IBH pathogenesis.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Aviadenovirus/fisiología , Pollos , Hepatitis Viral Animal/fisiopatología , Cuerpos de Inclusión Viral/fisiología , Pancreatitis/veterinaria , Enfermedades de las Aves de Corral/fisiopatología , Infecciones por Adenoviridae/metabolismo , Infecciones por Adenoviridae/fisiopatología , Infecciones por Adenoviridae/virología , Factores de Edad , Animales , Infecciones Asintomáticas , Digestión , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/fisiopatología , Susceptibilidad a Enfermedades/veterinaria , Susceptibilidad a Enfermedades/virología , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/virología , Pancreatitis/metabolismo , Pancreatitis/fisiopatología , Pancreatitis/virología , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/virología , Organismos Libres de Patógenos Específicos
10.
Mol Neurobiol ; 55(8): 6558-6571, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29327203

RESUMEN

Mouse hepatitis virus (MHV) infection causes meningoencephalitis by disrupting the neuro-glial and glial-pial homeostasis. Recent studies suggest that MHV infection alters gap junction protein connexin 43 (Cx43)-mediated intercellular communication in brain and primary cultured astrocytes. In addition to astrocytes, meningeal fibroblasts also express high levels of Cx43. Fibroblasts in the meninges together with the basal lamina and the astrocyte endfeet forms the glial limitans superficialis as part of the blood-brain barrier (BBB). Alteration of glial-pial gap junction intercellular communication (GJIC) in MHV infection has the potential to affect the integrity of BBB. Till date, it is not known if viral infection can modulate Cx43 expression and function in cells of the brain meninges and thus affect BBB permeability. In the present study, we have investigated the effect of MHV infection on Cx43 localization and function in mouse brain meningeal cells and primary meningeal fibroblasts. Our results show that MHV infection reduces total Cx43 levels and causes its intracellular retention in the perinuclear compartments reducing its surface expression. Reduced trafficking of Cx43 to the cell surface in MHV-infected cells is associated with loss functional GJIC. Together, these data suggest that MHV infection can directly affect expression and cellular distribution of Cx43 resulting in loss of Cx43-mediated GJIC in meningeal fibroblasts, which may be associated with altered BBB function observed in acute infection.


Asunto(s)
Conexina 43/deficiencia , Fibroblastos/patología , Fibroblastos/virología , Uniones Comunicantes/metabolismo , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/patología , Meninges/patología , Virus de la Hepatitis Murina/fisiología , Animales , Células Cultivadas , Conexina 43/metabolismo , Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Inflamación/patología , Ratones Endogámicos C57BL , Agregado de Proteínas , Vimentina/metabolismo
11.
Artículo en Inglés | MEDLINE | ID: mdl-28638862

RESUMEN

BACKGROUND: Duck viral hepatitis (DVH), caused by duck hepatitis A virus (DHAV), is a fatal contagious infectious disease which spreads rapidly with high morbidity and high mortality, and there is no effective clinical drug against DVH. MATERIALS AND METHODS: Raw Rehmannia Radix Polysaccharide (RRRP), Lycii Fructus polysaccharides and Astragalus Radix polysaccharides were experimented in vitro and in vivo. Mortality rate, livers change, liver lesion scoring, peroxidative injury evaluation indexes in vitro and in vivo, and hepatic injury evaluation indexes of optimal one were detected and observed in this experiment. RESULTS: RRRP could reduce mortality with the protection rate about 20.0% compared with that of the viral control (VC) group, finding that RRRP was the most effective against DHAV. The average liver scoring of the VC, blank control (BC), RRRP groups were 3.5, 0, 2.1. Significant difference (P<0.05) appeared between any two groups, demonstrating that it can alleviate liver pathological change. RRRP could make the hepatic injury evaluation indexes similar to BC group while the levels of the VC group were higher than other two groups in general. The levels of SOD, GSH-Px, CAT of RRRP group showed significant higher than that of VC group while the levels of NOS and MDA showed the opposite tendency, thus, RRRP could release peroxidative injury. CONCLUSION: RRRP was the most effective against duck hepatitis A virus (DHAV). RRRP could reduce mortality, alleviate liver pathological change, down-regulate liver lesion score, release peroxidative injury and hepatic injury. The antiviral and peroxidative injury releasing activity of RRRP for DHAV provided a platform to test novel drug strategies for hepatitis A virus in human beings.


Asunto(s)
Antivirales/administración & dosificación , Virus de la Hepatitis del Pato/efectos de los fármacos , Hepatitis Viral Animal/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Polisacáridos/administración & dosificación , Enfermedades de las Aves de Corral/tratamiento farmacológico , Rehmannia/química , Animales , Planta del Astrágalo/química , Patos , Virus de la Hepatitis del Pato/fisiología , Hepatitis Viral Animal/diagnóstico por imagen , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/virología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/virología , Raíces de Plantas/química , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/virología
12.
Hepatobiliary Pancreat Dis Int ; 16(3): 245-256, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28603092

RESUMEN

BACKGROUND: Liver inflammation or hepatitis is a result of pluripotent interactions of cell death molecules, cytokines, chemokines and the resident immune cells collectively called as microenvironment. The interplay of these inflammatory mediators and switching of immune responses during hepatotoxic, viral, drug-induced and immune cell-mediated hepatitis decide the fate of liver pathology. The present review aimed to describe the mechanisms of liver injury, its relevance to human liver pathology and insights for the future therapeutic interventions. DATA SOURCES: The data of mouse hepatic models and relevant human liver diseases presented in this review are systematically collected from PubMed, ScienceDirect and the Web of Science databases published in English. RESULTS: The hepatotoxic liver injury in mice induced by the metabolites of CCl4, acetaminophen or alcohol represent necrotic cell death with activation of cytochrome pathway, formation of reactive oxygen species (ROS) and mitochondrial damage. The Fas or TNF-alpha induced apoptotic liver injury was dependent on activation of caspases, release of cytochrome c and apoptosome formation. The ConA-hepatitis demonstrated the involvement of TRAIL-dependent necrotic/necroptotic cell death with activation of RIPK1/3. The alpha-GalCer-induced liver injury was mediated by TNF-alpha. The LPS-induced hepatitis involved TNF-alpha, Fas/FasL, and perforin/granzyme cell death pathways. The MHV3 or Poly(I:C) induced liver injury was mediated by natural killer cells and TNF-alpha signaling. The necrotic ischemia-reperfusion liver injury was mediated by hypoxia, ROS, and pro-inflammatory cytokines; however, necroptotic cell death was found in partial hepatectomy. The crucial role of immune cells and cell death mediators in viral hepatitis (HBV, HCV), drug-induced liver injury, non-alcoholic fatty liver disease and alcoholic liver disease in human were discussed. CONCLUSIONS: The mouse animal models of hepatitis provide a parallel approach for the study of human liver pathology. Blocking or stimulating the pathways associated with liver cell death could unveil the novel therapeutic strategies in the management of liver diseases.


Asunto(s)
Comunicación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Hepatitis Viral Animal/inmunología , Hígado/inmunología , Animales , Apoptosis , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteína Ligando Fas/metabolismo , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/patología , Hepatitis Viral Animal/virología , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Hígado/metabolismo , Hígado/patología , Hígado/virología , Ratones , Necrosis , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Especificidad de la Especie
13.
Mol Immunol ; 87: 171-179, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28494352

RESUMEN

Fulminant hepatitis (FH) is characterized by rapid liver failure and high mortality. The pathogenesis of viral FH includes virus-induced immune activation, inflammation, and subsequent hepatic apoptosis and necrosis. However, the mechanisms that underlie FH progression are unclear. IL-33 is a member of the IL-1-related cytokines, considered to be an "alarmin" that participates in various diseases, but its precise role in the coagulation of FH is not very clear. In our study, we found that IL-33 is significantly elevated in mice infected with murine hepatitis virus strain 3 (MHV-3). This is accompanied by an increase in pro-coagulant fibrinogen-like protein 2 (FGL2) in the liver. Previous studies have suggested that an increase in FGL2 is diagnostic of FH and liver necrosis, and animals with no FGL2 had better survivorship during FH. Our studies showed that IL-33 administration in a MHV-3 infection promoted survival during FH, with a significant reduction in FGL2 expression and liver inflammation. In vitro IL-33 treatment abrogated MHV-3 and IFN-γ induced FGL2 expression in RAW264.7 and THP-1 cells, respectively. In conclusion, our research suggests that IL-33 protects against viral fulminant hepatitis in mice by antagonizing expression of the pro-coagulant protein FGL2.


Asunto(s)
Coagulación Sanguínea/fisiología , Fibrinógeno/metabolismo , Hepatitis Viral Animal/metabolismo , Interleucina-33/metabolismo , Virus de la Hepatitis Murina/metabolismo , Animales , Apoptosis/fisiología , Femenino , Inflamación/metabolismo , Inflamación/virología , Interferón gamma/metabolismo , Interleucina-1/metabolismo , Hígado/metabolismo , Hígado/virología , Ratones , Ratones Endogámicos BALB C
14.
PLoS One ; 12(4): e0175495, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28394931

RESUMEN

The principal target organ of duck hepatitis A virus type 1 (DHAV-1) is duckling liver, which is an energy-intensive organ and plays important roles in body's energy metabolism and conversion. As the "power house" of the hepatocytes, mitochondria provide more than 90% of the energy. However, mitochondria are much vulnerable to the oxidative stress for their rich in polyunsaturated fatty acids. Although previous researches have demonstrated that DHAV-1 could induce the oxidative stress in the serum of the infected ducklings, no related study on the mitochondria during the pathological process of DVH has been reported by far. To address this issue, we examined the HE stained tissue pathological slices, detected the hepatic SOD, CAT and GPX activities and MDA contents and analyzed the ATP content, mitochondrial ultrastructure and the mitochondrial SOD, GPX activities and MDA content in the liver tissues. The results showed that the hepatic redox status was significantly disturbed so that causing the mitochondrial dysfunction, ATP depletion and mitochondrial oxidative stress during the process of the DHAV-1 infection, and a prescription formulated with Hypericum japonicum flavone, Radix Rehmanniae Recens polysaccharide and Salvia plebeia flavone (HRS), which had been demonstrated with good anti-oxidative activity in serum, could effectively alleviate the hepatic injury and the oxidative stress in liver tissue induced by DHAV-1 thus alleviating the mitochondrial injury and oxidative stress. In a word, this research discovers the oxidative stress induced mitochondrial dysfunction and oxidative stress during the DVH pathological process and demonstrates HRS exerts good anti-oxidative activity in liver tissue to protect mitochondria against reactive oxygen species (ROS).


Asunto(s)
Antivirales/farmacología , Medicamentos Herbarios Chinos/farmacología , Virus de la Hepatitis del Pato , Hepatitis Viral Animal/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Infecciones por Picornaviridae/tratamiento farmacológico , Adenosina Trifosfato/metabolismo , Animales , Patos , Flavonas/farmacología , Glutatión Peroxidasa/metabolismo , Hepatitis Viral Animal/metabolismo , Hepatitis Viral Animal/mortalidad , Hepatitis Viral Animal/patología , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Malondialdehído/metabolismo , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Estrés Oxidativo/efectos de los fármacos , Infecciones por Picornaviridae/metabolismo , Infecciones por Picornaviridae/mortalidad , Infecciones por Picornaviridae/patología , Polisacáridos/farmacología , Distribución Aleatoria , Superóxido Dismutasa/metabolismo , Resultado del Tratamiento
15.
Sci Rep ; 6: 28776, 2016 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-27354260

RESUMEN

IFI16 and AIM2 are important DNA sensors in antiviral immunity. To characterize these two molecules in a woodchuck model, which is widely used to study hepatitis B virus (HBV) infection, we cloned and analyzed the complete coding sequences (CDSs) of woodchuck IFI16 and AIM2, and found that AIM2 was highly conserved in mammals, whereas the degree of sequence identity between woodchuck IFI16 and its mammalian orthologues was low. IFI16 and IFN-ß were upregulated following VACV ds 70 mer transfection, while AIM2 and IL-1ß were upregulated following poly (dA:dT) transfection, both in vitro and in vivo; IFI16-targeted siRNA decreased the transcription of IFI16 and IFN-ß stimulated by VACV ds 70 mer, and AIM2 siRNA interference downregulated AIM2 and IL-1ß transcripts stimulated by poly (dA:dT), in vitro, suggesting that woodchuck IFI16 and AIM2 may play pivotal roles in the DNA-mediated induction of IFN-ß and IL-1ß, respectively. IFI16 and AIM2 transcripts were upregulated in the liver and spleen following acute WHV infection, while IFI16 was downregulated in the liver following chronic infection, implying that IFI16 and AIM2 may be involved in WHV infection. These data provide the basis for the study of IFI16- and AIM2-mediated innate immunity using the woodchuck model.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Virus de Hepatitis/inmunología , Hepatitis Viral Animal/metabolismo , Marmota/metabolismo , Enfermedades de los Roedores/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Secuencia Conservada , Proteínas de Unión al ADN/genética , Expresión Génica , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/virología , Interferón beta/genética , Interferón beta/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Hígado/metabolismo , Marmota/virología , Filogenia , Enfermedades de los Roedores/inmunología , Enfermedades de los Roedores/virología , Roedores , Bazo/metabolismo
16.
PLoS One ; 11(4): e0153723, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27073893

RESUMEN

The infection of chickens with avian Hepatitis E virus (avian HEV) can be asymptomatic or induces clinical signs characterized by increased mortality and decreased egg production in adult birds. Due to the lack of an efficient cell culture system for avian HEV, the interaction between virus and host cells is still barely understood. In this study, four truncated avian HEV capsid proteins (ORF2-1 - ORF2-4) with an identical 338aa deletion at the N-terminus and gradual deletions from 0, 42, 99 and 136aa at the C-terminus, respectively, were expressed and used to map the possible binding site within avian HEV capsid protein. Results from the binding assay showed that three truncated capsid proteins attached to avian LMH cells, but did not penetrate into cells. However, the shortest construct, ORF2-4, lost the capability of binding to cells suggesting that the presence of amino acids 471 to 507 of the capsid protein is crucial for the attachment. The construct ORF2-3 (aa339-507) was used to study the potential binding of avian HEV capsid protein to human and other avian species. It could be demonstrated that ORF2-3 was capable of binding to QT-35 cells from Japanese quail and human HepG2 cells but failed to bind to P815 cells. Additionally, chicken serum raised against ORF2-3 successfully blocked the binding to LMH cells. Treatment with heparin sodium salt or sodium chlorate significantly reduced binding of ORF2-3 to LMH cells. However, heparinase II treatment of LMH cells had no effect on binding of the ORF2-3 construct, suggesting a possible distinct attachment mechanism of avian as compared to human HEV. For the first time, interactions between avian HEV capsid protein and host cells were investigated demonstrating that aa471 to 507 of the capsid protein are needed to facilitate interaction with different kind of cells from different species.


Asunto(s)
Proteínas de la Cápside/metabolismo , Hepatitis Viral Animal/metabolismo , Hepevirus/metabolismo , Infecciones por Virus ARN/metabolismo , Animales , Pollos/virología , Hepatitis Viral Animal/virología , Interacciones Huésped-Patógeno , Humanos , Infecciones por Virus ARN/virología
17.
J Pineal Res ; 61(2): 168-76, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27101794

RESUMEN

The sphingosine kinase (SphK)1/sphingosine-1-phosphate (S1P) pathway is involved in multiple biological processes, including liver diseases. This study investigate whether modulation of the SphK1/S1P system associates to the beneficial effects of melatonin in an animal model of acute liver failure (ALF) induced by the rabbit hemorrhagic disease virus (RHDV). Rabbits were experimentally infected with 2 × 10(4) hemagglutination units of a RHDV isolate and received 20 mg/kg of melatonin at 0, 12, and 24 hr postinfection. Liver mRNA levels, protein concentration, and immunohistochemical labeling for SphK1 increased in RHDV-infected rabbits. S1P production and protein expression of the S1PR1 receptor were significantly elevated following RHDV infection. These effects were significantly reduced by melatonin. Rabbits also exhibited increased expression of toll-like receptor (TLR)4, tumor necrosis factor alpha (TNF-α), interleukin (IL)-6, nuclear factor-kappa B (NF-κB) p50 and p65 subunits, and phosphorylated inhibitor of kappa B (IκB)α. Melatonin administration significantly inhibited those changes and induced a decreased immunoreactivity for RHDV viral VP60 antigen in the liver. Results obtained indicate that the SphK1/S1P system activates in parallel to viral replication and the inflammatory process induced by the virus. Inhibition of the lipid signaling pathway by the indole reveals novel molecular pathways that may account for the protective effect of melatonin in this animal model of ALF, and supports the potential of melatonin as an antiviral agent.


Asunto(s)
Infecciones por Caliciviridae/metabolismo , Virus de la Enfermedad Hemorrágica del Conejo , Hepatitis Viral Animal/metabolismo , Fallo Hepático Agudo/metabolismo , Lisofosfolípidos/metabolismo , Melatonina/farmacocinética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Transducción de Señal/efectos de los fármacos , Esfingosina/análogos & derivados , Animales , Infecciones por Caliciviridae/tratamiento farmacológico , Hepatitis Viral Animal/tratamiento farmacológico , Fallo Hepático Agudo/tratamiento farmacológico , Masculino , Conejos , Esfingosina/metabolismo
18.
Immunol Res ; 64(2): 518-30, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26482053

RESUMEN

The underlying immune-mediated mechanisms involved in virus-induced severe hepatitis have not been well elucidated. In this study, we investigated the role of CD3(+)CD4(-)CD8(-) double-negative T (DN T) cells in the pathogenesis of fulminant viral hepatitis (FVH) induced by murine hepatitis virus strain 3 (MHV-3). After MHV-3 infection, the proportions of DN T cells increased significantly in BALB/cJ mice, and splenic DN T cells expressing high levels of CD69 were recruited by MHV-3-infected hepatocytes to the liver. Serum levels of alanine aminotransferase, aspartate aminotransferase and total bilirubin increased, accompanied by massive hepatocyte necrosis. These DN T cells were predominantly consisted of a TCRαß(+) subset expressing high levels of CD44 and did not produce cytokine except IL-2. Adoptive transfer of this subset of DN T cells to the MHV-3-infected mice resulted in an increase in murine fibrinogen-like protein 2 (mfgl2) expressions in association with massive fibrin deposition in the liver. Following MHV-3 infection, membrane mfgl2 expression and functional procoagulant activity increased remarkably in the DN T cells. Introduction of a recombinant adenovirus which encoded a microRNA specifically targeting mfgl2 gene (Ad-mfgl2-miRNA) in vivo significantly inhibited the hepatic expression of mfgl2 and improved survival in mice. However, under this condition, adoptive transfer of the DN T cells accelerated the disease progression and reversed the benefit from mfgl2 gene silence, leading to a 100 % death rate. Our results demonstrate that DN T cells contribute to the outcome of MHV-3-induced FVH via an important effector molecule mfgl2.


Asunto(s)
Fibrinógeno/metabolismo , Hepatitis Viral Animal/inmunología , Hepatitis Viral Animal/metabolismo , Virus de la Hepatitis Murina/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Traslado Adoptivo , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Fibrinógeno/genética , Silenciador del Gen , Hepatitis Viral Animal/mortalidad , Hepatitis Viral Animal/terapia , Inmunofenotipificación , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Activación de Linfocitos , Ratones , Fenotipo , ARN Interferente Pequeño/genética
19.
J Hepatol ; 62(2): 354-62, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25200905

RESUMEN

BACKGROUND & AIMS: Viral fulminant hepatitis (FH) is a disease with a high mortality rate. Activation of the complement system correlates with the development of FH. However, the key factors mediating complement activation in FH remain elusive. METHODS: Liver tissues were isolated from FH patients infected by hepatitis B virus (HBV) and from mice infected with murine hepatitis virus strain 3 (MHV-3). Wild type mice were treated with or without antagonists of C5aR or TNF-α, and mice deficient for C5aR (C5aR(-/-)), Fgl2 (Fgl2(-/-)), and Tnfα (Tnfα(-/-)) mice were not treated with the antagonists. C5b-9, C5aR, FGL2, CD31, CD11b, fibrin, TNF-α, and complement C3 cleavage products were detected by immunohistochemistry, immunofluorescence, or ELISA. Sorted liver sinusoidal endothelial cells (LSECs) or myeloid-derived (CD11b(+)) cells were stimulated with C5a, TNF-α or MHV-3 in vitro. The mRNA expressions levels of Fgl2 and Tnfα were determined by qRT-PCR analyses. RESULTS: We observed that complement activation, coagulation and pro-inflammatory cytokine production were upregulated in the HBV(+) patients with FH. Similar observations were made in the murine FH models. Complement activation and coagulation were significantly reduced in MHV-3 infected mice in the absence of C5aR, Tnfα or Fgl2. The MHV-3 infected C5aR(-/-) mice exhibited reduced numbers of infiltrated inflammatory CD11b(+) cells and a reduced expression of TNF-α and FGL2. Moreover, C5a administration stimulated TNF-α production by CD11b(+) cells, which in turn promoted the expression of FGL2 in CD31(+) LSEC-like cells in vitro. Administration of antagonists against C5aR or TNF-α ameliorated MHV-3-induced FH. CONCLUSIONS: Our results demonstrate that C5aR, TNF-α, and FGL2 form an integral network that contributes to coagulation and complement activation, and suggest that those are potential therapeutic targets in viral FH intervention.


Asunto(s)
Coagulación Sanguínea/genética , Activación de Complemento/genética , Fibrinógeno/genética , Regulación de la Expresión Génica , Hepatitis Viral Animal/metabolismo , Receptor de Anafilatoxina C5a/genética , Factor de Necrosis Tumoral alfa/genética , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrinógeno/biosíntesis , Hepatitis Viral Animal/patología , Hepatitis Viral Animal/virología , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/patogenicidad , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Anafilatoxina C5a/biosíntesis , Linfocitos T , Factor de Necrosis Tumoral alfa/biosíntesis
20.
Int J Biol Macromol ; 66: 186-93, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24582875

RESUMEN

In order to study the antioxidant effect of Bush Sophora Root polysaccharide (BSRPS) and its sulfate on anti-duck virus hepatitis (DVH), sulfated Bush Sophora Root polysaccharide (sBSRPS) was prepared by chlorosulfonic acid-pyridine method. Ducklings were fed with BSRPS and sBSRPS after challenged DHAV. Death was monitored, evaluation indexes of peroxidative and hepatic injury at the initial (4th and 8th hour) and later (54th hour) stages were detected. The results showed a fine treatment effect of BSRPS and sBSRPS. Visual hepatic pathological injury severities were less serious after the treatment. At the initial stage, free radical levels in all groups were the same, and BSRPS and sBSRPS reduced the hepatic injury through inhibiting virus replication. At the later stage, mass free radicals were detected in VC group while free radical levels in BSRPS and sBSRPS groups were significantly lower than VC group. The antioxidant effect of BSRPS and sBSRPS might alleviate the hepatic injury.


Asunto(s)
Depuradores de Radicales Libres/farmacología , Virus de la Hepatitis del Pato/efectos de los fármacos , Hepatitis Viral Animal/tratamiento farmacológico , Raíces de Plantas/química , Polisacáridos/farmacología , Sophora/química , Sulfatos/farmacología , Animales , Antioxidantes/química , Antioxidantes/farmacología , Antivirales/química , Antivirales/farmacología , ADN Viral/metabolismo , Patos/virología , Depuradores de Radicales Libres/química , Radicales Libres/metabolismo , Hepatitis Viral Animal/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/virología , Mortalidad , Infecciones por Picornaviridae/tratamiento farmacológico , Infecciones por Picornaviridae/metabolismo , Polisacáridos/química , Piridinas/química , Piridinas/farmacología , Sulfatos/química , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA