Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Viruses ; 13(8)2021 07 26.
Article in English | MEDLINE | ID: mdl-34452320

ABSTRACT

Since its first discovery by Arnold Theiler in 1918, serum hepatitis also known as Theiler's disease has been reported worldwide, causing idiopathic acute hepatitis and liver failure in horses. Recent studies have suggested a novel parvovirus, named equine parvovirus hepatitis (EqPV-H), to be associated with Theiler's disease. Despite the severity and potential fatality of EqPV-H infection, little is known about the possibility of developing chronic infections and putative cross-species infection of equine sister species. In the present longitudinal study, we employed qPCR analysis, serology, and biochemical testing as well as pathology examination of liver biopsies and sequence analysis to investigate potential chronic EqPV-H infection in an isolated study cohort of in total 124 horses from Germany over five years (2013-2018). Importantly, our data suggest that EqPV-H viremia can become chronic in infected horses that do not show biochemical and pathological signs of liver disease. Phylogenetic analysis by maximum likelihood model also confirms high sequence similarity and nucleotide conservation of the multidomain nuclear phosphoprotein NS1 sequences from equine serum samples collected between 2013-2018. Moreover, by examining human, zebra, and donkey sera for the presence of EqPV-H DNA and VP1 capsid protein antibodies, we found evidence for cross-species infection in donkey, but not to human and zebra. In conclusion, this study provides proof for the occurrence of persistent EqPV-H infection in asymptomatic horses and cross-species EqPV-H detection in donkeys.


Subject(s)
Hepatitis, Viral, Animal/blood , Hepatitis, Viral, Animal/physiopathology , Parvoviridae Infections/physiopathology , Parvoviridae Infections/veterinary , Parvovirus/genetics , Viremia/veterinary , Animals , Biopsy , Cohort Studies , DNA, Viral/genetics , Horse Diseases/virology , Horses , Liver/pathology , Liver/virology , Longitudinal Studies , Parvoviridae Infections/blood , Parvovirus/classification , Persistent Infection , Phylogeny
2.
Avian Dis ; 62(1): 57-64, 2018 03.
Article in English | MEDLINE | ID: mdl-29620455

ABSTRACT

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.


Subject(s)
Adenoviridae Infections/veterinary , Aviadenovirus/physiology , Chickens , Hepatitis, Viral, Animal/physiopathology , Inclusion Bodies, Viral/physiology , Pancreatitis/veterinary , Poultry Diseases/physiopathology , Adenoviridae Infections/metabolism , Adenoviridae Infections/physiopathology , Adenoviridae Infections/virology , Age Factors , Animals , Asymptomatic Infections , Digestion , Disease Susceptibility/metabolism , Disease Susceptibility/physiopathology , Disease Susceptibility/veterinary , Disease Susceptibility/virology , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/virology , Pancreatitis/metabolism , Pancreatitis/physiopathology , Pancreatitis/virology , Poultry Diseases/metabolism , Poultry Diseases/virology , Specific Pathogen-Free Organisms
3.
Arch Virol ; 163(5): 1187-1193, 2018 May.
Article in English | MEDLINE | ID: mdl-29387970

ABSTRACT

To establish an animal model for the newly identified Marmota Himalayana hepatovirus, MHHAV, so as to develop a better understanding of the infection of hepatitis A viruses. Five experimental woodchucks (Marmota monax) were inoculated intravenously with the purified MHHAV from wild woodchuck feces. One animal injected with PBS was defined as a control. Feces and blood were routinely collected. After the animals were subjected to necropsy, different tissues were collected. The presence of viral RNA and negative sense viral RNA was analyzed in all the samples and histopathological and in situ hybridization analysis was performed for the tissues. MHHAV infection caused fever but no severe symptoms or death. Virus was shed in feces beginning at 2 dpi, and MHHAV RNA persisted in feces for ~2 months, with a biphasic increase, and in blood for ~30 days. Viral RNA was detected in all the tissues, with high levels in the liver and spleen. Negative-strand viral RNA was detected only in the liver. Furthermore, the animals showed histological signs of hepatitis at 45 dpi. MHHAV can infect M. monax and is associated with hepatic disease. Therefore, this animal can be used as a model of HAV pathogenesis and to evaluate antiviral and anticancer therapeutics.


Subject(s)
Disease Models, Animal , Hepatitis A virus/pathogenicity , Hepatitis A , Hepatitis, Viral, Animal , Marmota , Animals , Feces/virology , Hepatitis A/pathology , Hepatitis A/physiopathology , Hepatitis A/virology , Hepatitis A virus/genetics , Hepatitis A virus/isolation & purification , Hepatitis A virus/physiology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/physiopathology , Hepatitis, Viral, Animal/virology , Liver/pathology , Liver/virology , RNA, Viral/isolation & purification , Spleen/pathology , Spleen/virology
4.
J Hepatol ; 68(4): 682-690, 2018 04.
Article in English | MEDLINE | ID: mdl-29274730

ABSTRACT

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.


Subject(s)
Hepatitis, Viral, Animal/physiopathology , Kupffer Cells/physiology , Receptor, Interferon alpha-beta/physiology , Signal Transduction/physiology , Acute Disease , Animals , Hepatitis, Viral, Animal/etiology , Mice , Mice, Inbred C57BL , Vaccinia/physiopathology
5.
Vet Microbiol ; 147(3-4): 310-9, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-20708350

ABSTRACT

A genetically distinct strain of avian hepatitis E virus (avian HEV-VA strain) was isolated from a healthy chicken in Virginia, and thus it is important to characterize and compare its pathogenicity with the prototype strain (avian HEV-prototype) isolated from a diseased chicken. Here we first constructed an infectious clone of the avian HEV-VA strain. Capped RNA transcripts from the avian HEV-VA clone were replication-competent after transfection of LMH chicken liver cells. Chickens inoculated intrahepatically with RNA transcripts of avian HEV-VA clone developed active infection as evidenced by fecal virus shedding, viremia, and seroconversion. To characterize the pathogenicity, RNA transcripts of both avian HEV-VA and avian HEV-prototype clones were intrahepatically inoculated into the livers of chickens. Avian HEV RNA was detected in feces, serum and bile samples from 10/10 avian HEV-VA-inoculated and 9/9 avian HEV-prototype-inoculated chickens although seroconversion occurred only in some chickens during the experimental period. The histopathological lesion scores were lower for avian HEV-VA group than avian HEV-prototype group in the liver at 3 and 5 weeks post-inoculation (wpi) and in the spleen at 3 wpi, although the differences were not statistically significant. The liver/body weight ratio, indicative of liver enlargement, of both avian HEV-VA and avian HEV-prototype groups were significantly higher than that of the control group at 5 wpi. Overall, the avian HEV-VA strain still induces histological liver lesions even though it was isolated from a healthy chicken. The results also showed that intrahepatic inoculation of chickens with RNA transcripts of avian HEV infectious clone may serve as an alternative for live virus in animal pathogenicity studies.


Subject(s)
DNA, Complementary/metabolism , Hepatitis E/veterinary , Hepatitis, Viral, Animal/virology , Hepevirus/pathogenicity , Poultry Diseases/virology , Animals , Cells, Cultured , Chickens , Cloning, Molecular , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Hepatitis E/pathology , Hepatitis E/virology , Hepatitis, Viral, Animal/physiopathology , Hepevirus/genetics , Liver/pathology , Liver/virology , Poultry Diseases/pathology , RNA Caps/genetics , Specific Pathogen-Free Organisms , Virginia , Virus Shedding
6.
J Gen Virol ; 91(Pt 2): 509-20, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19846670

ABSTRACT

The early phase after hepatitis B virus infection could play a crucial role in clearance and/or persistence of the virus, particularly in neonates. This work compared the early phase of duck hepatitis B virus infection in 1-day-old (D1) and 28-day-old (D28) ducks to determine whether differences in viral or host innate immune response can be related to the difference in outcome. In the first phase, almost immediately after inoculation, virus was taken up by components of the reticulo-endothelial systems, particularly liver-specific macrophages, Kupffer cells. Very early after infection, the induction of alpha interferon by infected hepatocytes occurred and was rapidly reinforced by recruitment of effector lymphocytes, which directly or indirectly caused apoptosis, eliminating infected hepatocytes, as was seen in mature birds. In addition, a lack of lymphocytic infiltration of the liver was found in D1 ducks, which supports the suggestion that the innate immune network is less effective in D1 ducks. Taken together, these results suggest that failure of the co-ordinated innate immune response rather than a defect in induced antiviral cell-mediated immunity may be the key factor which makes baby ducks vulnerable to persistence of hepadnavirus infection.


Subject(s)
Ducks , Hepadnaviridae Infections/veterinary , Hepatitis B Virus, Duck/immunology , Hepatitis, Viral, Animal/immunology , Poultry Diseases/immunology , Age Factors , Animals , Apoptosis , Cells, Cultured , Female , Hepadnaviridae Infections/immunology , Hepadnaviridae Infections/physiopathology , Hepadnaviridae Infections/virology , Hepatitis B Virus, Duck/isolation & purification , Hepatitis, Viral, Animal/physiopathology , Hepatitis, Viral, Animal/virology , Hepatocytes/cytology , Hepatocytes/immunology , Immunity, Cellular , Immunity, Innate , Lymphocytes/immunology , Male , Poultry Diseases/physiopathology , Poultry Diseases/virology
7.
Int Immunopharmacol ; 9(5): 627-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19258054

ABSTRACT

Mouse hepatitis virus strain A59 (MHV-A59) triggers various pathologies in several mouse strains, including hypergammaglobulinaemia, hepatitis and thymus involution. We reported previously the presence of autoantibodies (autoAb) to liver and kidney fumarylacetoacetate hydrolase (FAH) in sera from mice infected with MHV-A59. Long-term MHV-infected mice represented a good model of non-pathogenic autoimmune response since the animals were apparently healthy in spite of the presence of autoAb. The aim of this work was to see whether a severe liver injury, which releases endogenous adjuvants, i.e. danger signals, could elicit a broader spectrum of autoAb and perhaps signs of autoimmune hepatitis. Carbon tetrachloride (CCl(4)) was injected into mice 30 days after MHV infection, and serum was assayed for autoAb and total IgG 20 days later. The association of MHV infection with the toxic effects of CCl(4) resulted in hypergammaglobulinaemia and the production of autoAb to various liver and kidney proteins. Histological examination of liver samples showed tissue damages but without significant differences between the animals submitted to MHV+CCl(4) and controls, which were either infected by MHV without CCl(4), or poisoned by CCl(4) in the absence of MHV infection. Those results show that liver injury after viral infection may lead to the spreading of the immune response and to an increase of serum IgG, suggesting that the procedure used herein could simulate the onset of autoimmune hepatitis.


Subject(s)
Autoimmunity , Carbon Tetrachloride/toxicity , Hepatitis, Viral, Animal/immunology , Liver/immunology , Murine hepatitis virus/immunology , Animals , Autoantibodies/immunology , Female , Hepatitis, Autoimmune/etiology , Hepatitis, Viral, Animal/complications , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/physiopathology , Hydrolases/immunology , Kidney/drug effects , Kidney/immunology , Kidney/metabolism , Kidney/pathology , Kidney/virology , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver/virology , Mice , Mice, Inbred BALB C , Organ Specificity
8.
J Physiol Pharmacol ; 59 Suppl 1: 107-17, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18802219

ABSTRACT

The liver is the largest organ of the body. It is located between the portal and the general circulation, between the organs of the gastrointestinal tract and the heart. The main function of the liver is to take up nutrients, to store them, and to provide nutrients to the other organs. At the same time has the liver to take up potentially damaging substances like bacterial products or drugs delivered by the portal blood or microorganisms, which reach the circulation. The liver is not only an important power and sewage treatment plant of the body. In fact, the liver is probably the best example for a cheap recycling system. Both parenchymal and nonparenchymal liver cells participate in the clearance activities. The function of the liver as clearance organ, however, harbors the danger that the substances that should be degraded and/or eliminated lead to tissue damage. Thus, effective defense mechanisms are necessary. Among the nonparenchymal cells Kupffer cells, sinusoidal endothelial cells, and natural killer (NK) lymphocytes exert cellular defense functions for the whole body but also for the liver itself. Furthermore, each cell type of the liver, including the hepatocytes, possesses its own defense apparatus.


Subject(s)
Hepatitis/pathology , Hepatitis/physiopathology , Liver Regeneration , Liver/pathology , Liver/physiopathology , Animals , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/physiopathology , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/physiopathology , Gamma Rays , Hepatitis/etiology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/physiopathology , Hepatocytes/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Rats
9.
Virology ; 362(1): 10-5, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17412386

ABSTRACT

Rift Valley fever virus is a significant human and veterinary pathogen responsible for explosive outbreaks throughout Africa and the Arabian Peninsula. Severe acute disease in humans includes rapid onset hepatic disease and hemorrhagic fever or delayed onset encephalitis. A highly efficient reverse genetics system was developed which allowed generation of recombinant RVF viruses to assess the role of NSm protein in virulence in a rat model in which wild-type RVF virus strain ZH501 (wt-ZH501) results in 100% lethal hepatic disease 2-3 days post infection. While extensive genomic analysis indicates conservation of the NSm coding capability of diverse RVF viruses, and viruses deficient in NSs proteins are completely attenuated in vivo, comparison of wt-ZH501, a reverse genetics generated wt-ZH501 virus (R-ZH501), and R-ZH501 virus lacking the NSm proteins (R-DeltaNSm-ZH501) demonstrated that the NSm proteins were nonessential for in vivo virulence and lethality. Surprisingly, while 44% of R-DeltaNSm-ZH501 infected animals quickly developed lethal hepatic disease similar to wt- and R-ZH501, 17% developed delayed onset neurologic disease (lethargy, head tremors, and ataxia) at 13 days post infection. Such infections may provide the basis for study of both RVF acute hepatic disease and delayed onset encephalitic disease in humans.


Subject(s)
Encephalitis, Viral/virology , Rift Valley Fever/virology , Rift Valley fever virus/pathogenicity , Viral Nonstructural Proteins/genetics , Animals , Ataxia , Disease Models, Animal , Encephalitis, Viral/physiopathology , Female , Gene Deletion , Hepatitis, Viral, Animal/physiopathology , Hepatitis, Viral, Animal/virology , Lethargy , Rats , Rats, Inbred WF , Rift Valley Fever/physiopathology , Rift Valley fever virus/genetics , Survival Analysis , Tremor , Viral Nonstructural Proteins/physiology , Virulence/genetics
10.
Nutr. hosp ; 22(2): 199-209, mar.-abr. 2007. tab
Article in Es | IBECS | ID: ibc-055087

ABSTRACT

El fallo hepático fulminante (FHF) es un síndrome clínico muy grave, asociado con alta mortalidad, a pesar de los grandes avances que se han producido en los últimos años en la terapia tanto del manejo de los cuidados intensivos mediante diversos soportes hepáticos bioartificiales como del trasplante hepático. Tanto el conocimiento como el tratamiento del FHF han estado limitados por la falta de modelos animales satisfactorios. Así, han sido muchos los intentos de desarrollar un modelo adecuado, entre los que se incluyen los modelos quirúrgicos, tales como la hepatectomía y desvascularización total y/o parcial, la utilización de sustancias químicas con toxicidad hepática tales como el acetaminofeno, azoximetano, galactosamina, tioacetamida, entre otras. Ahora bien, la mayor parte de estos modelos no reflejan de modo idóneo el patrón de la enfermedad humana de FHF y todos ellos presentan importantes limitaciones. A pesar de que la hepatitis vírica es una de las etiologías más frecuentes de FHF, el uso de agentes víricos para desarrollar modelos animales ha sido escaso y desafortunado. Nuestro grupo ha desarrollado recientemente un modelo animal mediante la inoculación de conejos con el virus de la enfermedad hemorrágica del conejo que presenta características bioquímicas, histológicas y signos clínicos compatibles con el FHF del hombre. En el trabajo se resumen los modelos animales más utilizados asi como las ventajas e inconvenientes más reseñables de cada uno de ellos


Fulminant hepatic failure (FHF) is a very serious clinical síndrome that, in spite of the important therapeutical advances that have taken place in the last years by means of bioartifical hepatic support devices and hepatic transplantation, is still associated to a high mortality. Knowledge and treatment of the FHF have been limited by the lack of satisfactory animal models. Among the attempts to develop a suitable model are surgical models, such as hepatectomy and total and/or partial devascularization, or the use of chemical substances with hepatic toxicity, such as acetaminophen, azoximethane, galactosamine or thioacetamide, among others. However, most of these models do not adequatly reflect the pattern of the human disease and all of them present important limitations. Although viral hepatitis is one of the most frequent causes of FHF, the use of viral agents to develop animal models has been little and unfortunate. Our group has recently developed a viral animal model of FHF by means of the inoculation of rabbits with the virus of the rabbit hemorrhagic disease. This model displays biochemical, and histological characteristics, and clinical signs that ressemble those in human FHF. In the present article, the most widely used animal models of FHF, together with their main advantages and disadvantages, are presented


Subject(s)
Animals , Hepatic Insufficiency/physiopathology , Disease Models, Animal , Hemorrhagic Disease Virus, Rabbit/pathogenicity , Hepatitis, Viral, Animal/physiopathology , Hepatitis, Viral, Human/physiopathology , Acetaminophen/adverse effects , Models, Chemical , Models, Anatomic , Models, Biological
11.
World J Gastroenterol ; 12(47): 7626-34, 2006 Dec 21.
Article in English | MEDLINE | ID: mdl-17171791

ABSTRACT

AIM: To study the action of hepatitis virus infection-associated genes at transcription level during liver regeneration (LR). METHODS: Hepatitis virus infection-associated genes were obtained by collecting the data from databases and retrieving the correlated articles, and their expression changes in the regenerating rat liver were detected with the rat genome 230 2.0 array. RESULTS: Eighty-eight genes were found to be associated with liver regeneration. The number of genes initially and totally expressed during initial LR [0.5-4 h after partial hepatectomy (PH)], transition from G0 to G1 (4-6 h after PH), cell proliferation (6-66 h after PH), cell differentiation and reorganization of structure-function (66-168 h after PH) was 37, 8, 48, 3 and 37, 26, 80, 57, respectively, indicating that the genes were mainly triggered at the early stage of LR (0.5-4 h after PH), and worked at different phases. These genes were classified into 5 types according to their expression similarity, namely 37 up-regulated, 9 predominantly up-regulated, 34 down-regulated, 6 predominantly down-regulated and 2 up/down-regulated genes. Their total up- and down-regulation frequencies were 359 and 149 during LR, indicating that the expression of most genes was enhanced, while the expression of a small number of genes was attenuated during LR. According to time relevance, they were classified into 12 groups (0.5 and 1 h, 2 and 4 h, 6 h, 8 and 12 h, 16 and 96 h, 18 and 24 h, 30 and 42 h, 36 and 48 h, 54 and 60 h, 66 and 72 h, 120 and 144 h, 168 h), demonstrating that the cellular physiological and biochemical activities during LR were fluctuated. According to expression changes of the genes, their expression patterns were classified into 23 types, suggesting that the cellular physiological and biochemical activities during LR were diverse and complicated. CONCLUSION: The anti-virus infection capacity of regenerating liver can be enhanced and 88 genes play an important role in LR.


Subject(s)
Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/physiopathology , Liver Regeneration/genetics , Oligonucleotide Array Sequence Analysis , Animals , Female , Liver/physiology , Liver/virology , Male , Rats , Rats, Sprague-Dawley
12.
J Immunol ; 177(4): 2039-45, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16887960

ABSTRACT

The chemokine receptor CCR5 came into worldwide prominence a decade ago when it was identified as one of the major coreceptors for HIV infectivity. However, subsequent studies suggested an important modulatory role for CCR5 in the inflammatory response. Specifically, CCR5 has been reported to directly regulate T cell function in autoimmune diseases, including multiple sclerosis, rheumatoid arthritis, and type 1 diabetes. Moreover, T cell-mediated immune responses are proposed to be critical in the pathogenesis of autoimmune and viral liver diseases, and recent clinical and experimental studies have also implicated CCR5 in the pathogenesis of autoimmune and viral liver diseases. Therefore, in this brief review, we highlight the evidence that supports an important role of CCR5 in the pathophysiology of T cell-mediated liver diseases with specific emphasis on autoimmune and viral liver diseases.


Subject(s)
Liver Diseases/immunology , Liver Diseases/metabolism , Receptors, CCR5/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/metabolism , Hepatitis, Autoimmune/physiopathology , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/physiopathology , Hepatitis, Viral, Human/immunology , Hepatitis, Viral, Human/metabolism , Hepatitis, Viral, Human/physiopathology , Humans , Liver Diseases/physiopathology , T-Lymphocytes/pathology
13.
Anat Sci Int ; 80(4): 199-211, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16333916

ABSTRACT

Massive liver cell death provoked in silica-treated mice subsequently infected with herpes simplex virus (HSV)-1 is very similar pathohistologically to the cell death observed in human fulminant hepatitis. Previously, we have shown this liver cell death to be extensive apoptosis. In the present study, we examined various factors related to liver damage patho- and immunologically, as well as by reverse transcription-polymerase chain reaction. Tumor necrosis factor (TNF)-alpha, inducible nitric oxide synthase (iNOS), interferon (IFN)-alpha, and interleukin-6 mRNAs were detected to a much greater extent in silica-treated mice compared with control mice after HSV-1 infection, and excessive expression of iNOS mRNA and cytokine mRNAs in the liver may be closely related to massive liver cell apoptosis. The apoptosis was less related to the fas ligand than to TNF-alpha. Silica blockage of macrophages makes the liver cell extremely vulnerable to HSV-1 infection, and it induced expression of E-selectin and neutrophil margination in the liver. Subsequent HSV-1 infection induced excessive production of iNOS and cytokines, particularly TNF-alpha, but administration of anti-TNF-alpha antibody or NG-monomethyl-L-arginine was not completely efficacious for the survival of the mice. Overproduction of free radicals in combination with cytokines, such as TNF-alpha, IL-6 and IFN-alpha, may result in hepatic cell apoptosis.


Subject(s)
Apoptosis/physiology , Hepatitis, Viral, Animal/physiopathology , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Simplexvirus , Tumor Necrosis Factor-alpha/metabolism , Animals , DNA Primers , E-Selectin/metabolism , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Interferon-alpha/metabolism , Interleukin-6/metabolism , Liver/pathology , Liver/virology , Macrophages/drug effects , Mice , Mice, Inbred C3H , Reverse Transcriptase Polymerase Chain Reaction , Silicon Dioxide/toxicity
14.
J Gen Virol ; 86(Pt 9): 2585-2593, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16099918

ABSTRACT

Hepatitis E virus (HEV), the causative agent of hepatitis E, is an important human pathogen. Increasing evidence indicates that hepatitis E is a zoonosis. Avian HEV was recently discovered in chickens with hepatitis-splenomegaly syndrome in the USA. Like swine HEV from pigs, avian HEV is also genetically and antigenically related to human HEV. The objective of this study was to construct and characterize an infectious cDNA clone of avian HEV for future studies of HEV replication and pathogenesis. Three full-length cDNA clones of avian HEV, pT7-aHEV-5, pT7G-aHEV-10 and pT7G-aHEV-6, were constructed and their infectivity was tested by in vitro transfection of leghorn male hepatoma (LMH) chicken liver cells and by direct intrahepatic inoculation of specific-pathogen-free (SPF) chickens with capped RNA transcripts from the three clones. The results showed that the capped RNA transcripts from each of the three clones were replication competent when transfected into LMH cells as demonstrated by detection of viral antigens with avian HEV-specific antibodies. SPF chickens intrahepatically inoculated with the capped RNA transcripts from each of the three clones developed active avian HEV infections as evidenced by seroconversion to avian HEV antibodies, viraemia and faecal virus shedding. The infectivity was further confirmed by successful infection of naïve chickens with the viruses recovered from chickens inoculated with the RNA transcripts. The results indicated that all three cDNA clones of avian HEV are infectious both in vitro and in vivo. The availability of these infectious clones for a chicken strain of HEV now affords an opportunity to study the mechanisms of HEV cross-species infection and tissue tropism by constructing chimeric viruses among human, swine and avian HEVs.


Subject(s)
Cloning, Molecular , DNA, Complementary/genetics , Hepatitis E virus/pathogenicity , Hepatitis E/veterinary , Hepatitis, Viral, Animal/physiopathology , Poultry Diseases/physiopathology , Animals , Cell Line, Tumor , Chickens , DNA, Complementary/metabolism , Feces/virology , Hepatitis E/physiopathology , Hepatitis E/virology , Hepatitis E virus/classification , Hepatitis E virus/genetics , Hepatitis, Viral, Animal/virology , Poultry Diseases/virology , RNA Caps/genetics , RNA Caps/metabolism , Transcription, Genetic , Virus Replication
15.
J Virol ; 79(12): 7629-40, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15919915

ABSTRACT

Murine coronavirus A59 strain causes mild to moderate hepatitis in mice. We have previously shown that mutants of A59, unable to induce hepatitis, may be selected by persistent infection of primary glial cells in vitro. These in vitro isolated mutants encoded two amino acids substitutions in the spike (S) gene: Q159L lies in the putative receptor binding domain of S, and H716D, within the cleavage signal of S. Here, we show that hepatotropic revertant variants may be selected from these in vitro isolated mutants (Q159L-H716D) by multiple passages in the mouse liver. One of these mutants, hr2, was chosen for more in-depth study based on a more hepatovirulent phenotype. The S gene of hr2 (Q159L-R654H-H716D-E1035D) differed from the in vitro isolates (Q159L-H716D) in only 2 amino acids (R654H and E1035D). Using targeted RNA recombination, we have constructed isogenic recombinant MHV-A59 viruses differing only in these specific amino acids in S (Q159L-R654H-H716D-E1035D). We demonstrate that specific amino acid substitutions within the spike gene of the hr2 isolate determine the ability of the virus to cause lethal hepatitis and replicate to significantly higher titers in the liver compared to wild-type A59. Our results provide compelling evidence of the ability of coronaviruses to rapidly evolve in vivo to highly virulent phenotypes by functional compensation of a detrimental amino acid substitution in the receptor binding domain of the spike glycoprotein.


Subject(s)
Amino Acid Substitution , Evolution, Molecular , Hepatitis, Viral, Animal/physiopathology , Membrane Glycoproteins/chemistry , Murine hepatitis virus/pathogenicity , Receptors, Virus/metabolism , Viral Envelope Proteins/chemistry , Animals , Coronavirus Infections/pathology , Coronavirus Infections/physiopathology , Coronavirus Infections/virology , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Liver/pathology , Liver/virology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Murine hepatitis virus/genetics , Recombination, Genetic , Specific Pathogen-Free Organisms , Spike Glycoprotein, Coronavirus , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virulence
16.
Hepatology ; 41(5): 986-94, 2005 May.
Article in English | MEDLINE | ID: mdl-15793797

ABSTRACT

Only humans and chimpanzees are fully permissive for replication of hepatitis C virus (HCV), an important cause of liver cirrhosis and cancer worldwide. The absence of suitable animal models limits opportunities for in vivo evaluation of candidate hepatitis C therapeutics and slows progress in the field. Here, we describe a chimeric virus derived from GB virus B (GBV-B), an unclassified hepatotropic member of the family Flaviviridae that is closely related to HCV and infects tamarins (Saguinus sp.), in which a functionally important HCV regulatory sequence replaced an analogous sequence in the 5' nontranslated region (5'NTR) of the GBV-B genome. The transplanted sequence comprised domain III of the internal ribosome entry site (IRES), which directly binds the 40S ribosome subunit and is a target for candidate therapeutics. The chimeric 5'NTR retained ribosome binding activity and was competent in directing protein translation both in cell-free translation reactions and in transfected primary tamarin hepatocyte cultures. Virus rescued from the chimeric RNA replicated in the liver of tamarins, causing biochemical and histopathological changes typical of viral hepatitis. However, adaptive mutations were required elsewhere in the genome for efficient replication. Virus was not rescued from other, translationally competent, chimeric RNAs in which domain II of the IRES was exchanged. Thus, the 5'NTR appears to contain virus-specific replication signals that interact with other sites within the viral genome or with viral proteins. In conclusion, such novel chimeric flaviviruses offer opportunities for new insights into HCV replication mechanisms, while potentially facilitating the evaluation of candidate therapeutics in vivo.


Subject(s)
Disease Models, Animal , Flaviviridae Infections/physiopathology , GB virus B/genetics , Hepacivirus/genetics , Hepatitis, Viral, Animal/physiopathology , Saguinus/virology , 5' Untranslated Regions/genetics , Animals , Chimera , DNA, Complementary , Flaviviridae Infections/virology , GB virus B/growth & development , Genome, Viral , Hepatitis, Viral, Animal/virology , Plasmids/genetics , RNA, Viral/genetics , Ribosomes/genetics , Ribosomes/virology
17.
Hepatology ; 41(2): 280-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15660391

ABSTRACT

Inhibition of NFkappaB enhances the susceptibility of cancer to TRAIL-mediated apoptosis and is suggested as a strategy for cancer therapy. Because the role of NFkappaB in TRAIL-mediated apoptosis of hepatocytes is unknown, we investigated the influence of NFkappaB-inhibition in death ligand-mediated apoptosis in hepatitis. Adenoviral hepatitis resulted in upregulation of NFkappaB-activity, which could be inhibited by expression of IkappaBalpha-superrepressor. We treated mice after the onset of adenoviral hepatitis with adenoviruses expressing FasL (AdFasL), TRAIL (AdTRAIL), or GFP (AdGFP). In contrast to apoptosis induced by AdFasL, NFkappaB inhibition strongly enhanced AdTRAIL-mediated apoptosis of hepatocytes. Expression of IkappaBalpha inhibits adenoviral infection-mediated overexpression of bcl-xl, providing a molecular mechanism for TRAIL sensitization. In agreement with this hypothesis, downregulation of bcl-xl by siRNA enhanced susceptibility of hepatocytes to TRAIL, but not to FasL-mediated apoptosis, resulting in TRAIL-mediated severe liver damage after AdTRAIL application. Our data demonstrate that inhibition of NFkappaB in adenoviral hepatitis strongly sensitizes hepatocytes to TRAIL-mediated apoptosis. Bcl-xl, in contrast to bcl-2 and c-FLIP, is strongly upregulated after viral infection and represents an essential NFkappaB-dependent survival factor against TRAIL-mediated apoptosis. In conclusion, inhibition of NFkappaB or bcl-xl during TRAIL therapy may harbor a risk of liver damage in patients with viral hepatitis.


Subject(s)
Apoptosis , Hepatitis, Viral, Animal/physiopathology , Membrane Glycoproteins/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Animals , Apoptosis Regulatory Proteins , Cell Line , Fas Ligand Protein , Hepatitis, Viral, Animal/metabolism , Humans , Mice , Mice, Inbred BALB C , TNF-Related Apoptosis-Inducing Ligand , bcl-X Protein
18.
World J Gastroenterol ; 11(44): 6936-40, 2005 Nov 28.
Article in English | MEDLINE | ID: mdl-16437596

ABSTRACT

AIM: To evaluate the expression of fibrinogen-like protein 2 (fgl2) and its correlation with disease progression in both mice and patients with severe viral hepatitis. METHODS: Balb/cJ or A/J mice were infected intraperitoneally (ip) with 100 PFU of murine hepatitis virus type 3 (MHV-3), liver and serum were harvested at 24, 48, and 72 h post infection for further use. Liver tissues were obtained from 23 patients with severe acute chronic (AOC) hepatitis B and 13 patients with mild chronic hepatitis B. Fourteen patients with mild chronic hepatitis B with cirrhosis and 4 liver donors served as normal controls. In addition, peripheral blood mononuclear cells (PBMC) were isolated from 30 patients (unpaired) with severe AOC hepatitis B and 10 healthy volunteers as controls. Procoagulant activity representing functional prothrombinase activity in PBMC and white blood cells was also assayed. A polyclonal antibody against fgl2 was used to detect the expression of both mouse and human fgl2 protein in liver samples as well as in PBMC by immunohistochemistry staining in a separate set of studies. Alanine aminotransferase (ALT) and total bilirubin (TBil) in serum were measured to assess the severity of liver injury. RESULTS: Histological changes were found in liver sections 12-24 h post MHV-3 infection in Balb/cJ mice. In association with changes in liver histology, marked elevations in serum ALT and TBil were observed. Mouse fgl2 (mfgl2) protein was detected in the endothelium of intrahepatic veins and hepatic sinusoids within the liver 24 h after MHV-3 infection. Liver tissues from the patients with severe AOC hepatitis B had classical pathological features of acute necroinflammation. Human fgl2 (hfgl2) was detected in 21 of 23 patients (91.30%) with severe AOC hepatitis B, while only 1 of 13 patients (7.69%) with mild chronic hepatitis B and cirrhosis had hfgl2 mRNA or protein expression. Twenty-eight of thirty patients (93.33%) with severe AOC hepatitis B and 1 of 10 with mild chronic hepatitis B had detectable hfgl2 expression in PBMC. No hfgl2 expression was found either in the liver tissue or in the PBMC from normal donors. There was a positive correlation between hfgl2 expression and the severity of the liver disease as indicated by the levels of TBil. PCA significantly increased in PBMC in patients with severe AOC hepatitis B. CONCLUSION: The molecular and cellular results reported here in both mice and patients with severe viral hepatitis suggest that virus-induced hfgl2 prothrombinase/fibroleukin expression and the coagulation activity associated with the encoded fgl2 protein play a pivotal role in initiating severe hepatitis. The measurement of hfgl2/fibroleukin expression in PBMC may serve as a useful marker to monitor the severity of AOC hepatitis B and a target for therapeutic intervention.


Subject(s)
Fibrinogen/metabolism , Hepatitis B/physiopathology , Hepatitis, Viral, Animal/physiopathology , Murine hepatitis virus/metabolism , Animals , Disease Models, Animal , Disease Progression , Female , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatitis, Viral, Animal/metabolism , Hepatitis, Viral, Animal/pathology , Humans , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred BALB C , Murine hepatitis virus/genetics , Thromboplastin/metabolism
19.
Arch Virol ; 149(12): 2319-36, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15338320

ABSTRACT

Lymphocytic chorimeningitis virus (LCMV), the prototype arenavirus, and Lassa virus (LASV), causative agent of Lassa hemorrhagic fever (LHF), belong to the Old World group of the family Arenaviridae. Both viruses have extensive strain diversity and significant variations in lethality and pathogenicity for man and experimental animals. We have shown that the LHF-like infection of rhesus macaques with the WE strain of LCMV affects liver functions, induces hepatocyte proliferation, and causes a rise in IL-6 and soluble TNF receptors (sTNFR) concomitant with a rise in viremia. The levels of IL-6 and sTNFR can serve as an additional diagnostic tool for liver involvement in pathogenesis of arenavirus infection. Mucosal inoculation of rhesus macaques with LCMV-WE can result in attenuated infection with a transient viremia and liver enzyme abnormalities. The ARM strain of LCMV shares 88% amino acid homology with WE. In contrast to LCMV-WE, ARM strain does not induce manifested disease in monkeys, does not affect liver functions, and does not induce hepatocyte proliferation. Previously we demonstrated that LCMV-ARM infection protected rhesus macaques challenged with LCMV-WE. Here we have shown that the protected animals have no signs of hepatitis and hepatocyte proliferation.


Subject(s)
Arenaviridae Infections/physiopathology , Hepatitis, Viral, Animal/physiopathology , Hepatocytes/virology , Liver Regeneration/physiology , Lymphocytic choriomeningitis virus/pathogenicity , Animals , Arenaviridae Infections/immunology , Hepatitis, Viral, Animal/immunology , Hepatitis, Viral, Animal/virology , Interleukin-6/blood , Ki-67 Antigen/blood , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Macaca mulatta , Receptors, Tumor Necrosis Factor/blood , Species Specificity , Time Factors , Viremia/immunology , Virulence
20.
J Virol ; 78(4): 2062-71, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14747571

ABSTRACT

GB virus B (GBV-B), a flavivirus closely related to HCV, has previously been shown to infect and replicate to high titers in tamarins (Saguinus sp.). This study describes the use of GBV-B infection and replication in the common marmoset (Callithrix jacchus) for the successful development and validation of a surrogate animal model for hepatitis C virus (HCV). Infection of marmosets with GBV-B produced a viremia that peaked at 10(8) to 10(9) genome copies/ml for a period of 40 to 60 days followed by viral clearance at 60 to 80 days postinfection. Passage of the initial tamarin-derived GBV-B in marmosets produced an infectious stock that gave a more reproducible and consistent infection in the marmoset. Titration of the virus stocks in vivo indicated that they contained 1 infectious unit for every 1,000 genome copies. Cultures of primary marmoset hepatocytes were also successfully infected with GBV-B, with high levels of virus detected in supernatants and cells for up to 14 days postinfection. Treatment of GBV-B-infected hepatocyte cultures with a novel class of HCV protease inhibitor (pyrrolidine 5,5 trans-lactams) reduced viral levels by more than 2 logs. Treatment of GBV-B-infected marmosets with one such inhibitor resulted in a 3-log drop in serum viral titer over 4 days of therapy. These studies provide the first demonstration of the in vivo efficacy of a small-molecule inhibitor for HCV in an animal model and illustrate the utility of GBV-B as a surrogate animal model system for HCV.


Subject(s)
Callithrix/virology , Disease Models, Animal , Flaviviridae Infections/veterinary , GB virus B/pathogenicity , Hepatitis, Viral, Animal/physiopathology , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Flaviviridae Infections/drug therapy , Flaviviridae Infections/physiopathology , Flaviviridae Infections/virology , GB virus B/drug effects , Hepatitis C/drug therapy , Hepatitis C/physiopathology , Hepatitis C/virology , Hepatitis, Viral, Animal/drug therapy , Hepatitis, Viral, Animal/virology , Hepatocytes/virology , Humans , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Saguinus/virology , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...