Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 6: 39479, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27995987

ABSTRACT

Sunlight has important biological effects in human skin. Ultraviolet (UV) light striking the epidermis catalyzes the synthesis of Vitamin D and triggers melanin production. Although a causative element in skin cancers, sunlight is also associated with positive health outcomes including reduced incidences of autoimmune diseases and cancers. The mechanisms, however, by which light affects immune function remain unclear. Here we describe direct photon sensing in human and mouse T lymphocytes, a cell-type highly abundant in skin. Blue light irradiation at low doses (<300 mJ cm-2) triggers synthesis of hydrogen peroxide (H2O2) in T cells revealed by the genetically encoded reporter HyPerRed. In turn, H2O2 activates a Src kinase/phospholipase C-γ1 (PLC-γ1) signaling pathway and Ca2+ mobilization. Pharmacologic inhibition or genetic disruption of Lck kinase, PLC-γ1 or the T cell receptor complex inhibits light-evoked Ca2+ transients. Notably, both light and H2O2 enhance T-cell motility in a Lck-dependent manner. Thus, T lymphocytes possess intrinsic photosensitivity and this property may enhance their motility in skin.


Subject(s)
Cell Movement/radiation effects , Skin/radiation effects , T-Lymphocytes/cytology , T-Lymphocytes/radiation effects , Animals , Calcium/chemistry , Cell Proliferation , Chemotaxis , Humans , Hydrogen Peroxide , Jurkat Cells , Mice , Phospholipase C gamma/metabolism , Phosphorylation , Photons , RNA Interference , Signal Transduction/drug effects , Spleen/cytology , Sunlight , Ultraviolet Rays
2.
Alcohol Clin Exp Res ; 30(9): 1615-23, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16930225

ABSTRACT

BACKGROUND: Chronic alcohol drinking accelerates the progression of liver disease in patients with hepatitis viral infection; however, the underlying mechanisms are not fully understood. METHODS: Here, we examined the effects of chronic ethanol feeding on hepatic natural killer (NK) cells and liver injury in 2 murine models of liver injury: injection of synthetic double-stranded RNA polyinosinic-polycytidylic acid (poly I:C), which mimics viral infection, and infection with murine cytomegalovirus (MCMV). Mice were fed the Lieber-DeCarli liquid diet containing 5% (vol/vol) ethanol for 8 weeks, resulting in a significant decrease in the percentage and total number of NK cells in the liver. RESULTS: In control, pair-fed mice, poly I:C injection induced NK cell accumulation in the liver and activated hepatic NK cell cytotoxicity, whereas such induction and activation were diminished in ethanol-fed mice. Treatment with poly I:C also induced expression of NKG2D, granzyme B, perforin, Fas L, TRAIL, and IFN-gamma on liver lymphocytes, which were delayed or reduced in ethanol-treated mice compared with pair-fed mice. In contrast, chronic ethanol feeding did not affect poly I:C-induced mild liver injury. Furthermore, MCMV infection activated hepatic NK cells and induced hepatic inflammation and injury. Chronic ethanol consumption inhibited hepatic NK cell activation during MCMV infection, but enhanced MCMV-induced liver injury, viral titer, and inflammation in the liver. CONCLUSIONS: Taken together, these findings suggest that chronic ethanol consumption decreases hepatic NK activity, thereby accelerating MCMV-induced hepatitis and liver injury.


Subject(s)
Central Nervous System Depressants/pharmacology , Cytomegalovirus Infections/pathology , Ethanol/pharmacology , Hepatitis, Viral, Animal/pathology , Killer Cells, Natural/drug effects , Liver/cytology , Alanine Transaminase/blood , Animals , Antiviral Agents/pharmacology , Aspartate Aminotransferases/blood , Flow Cytometry , Liver/drug effects , Liver/pathology , Lymphocyte Count , Male , Mice , Mice, Inbred C57BL , Poly I-C/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
3.
Gastroenterology ; 130(2): 435-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16472598

ABSTRACT

BACKGROUND & AIMS: Viral hepatitis infection, which is a major cause of liver fibrosis, is associated with activation of innate immunity. However, the role of innate immunity in liver fibrosis remains obscure. METHODS: Liver fibrosis was induced either by feeding mice with the 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet or by injecting them with carbon tetrachloride. The Toll-like receptor 3 ligand, polyinosinic-polycytidylic acid, was used to activate innate immunity cells and mediators, including natural killer cells and interferon gamma. RESULTS: In the mouse model of DDC-induced liver fibrosis, natural killer cell activation by polyinosinic-polycytidylic acid induced cell death to activated hepatic stellate cells and attenuated the severity of liver fibrosis. Polyinosinic-polycytidylic acid treatment also ameliorated liver fibrosis induced by carbon tetrachloride. The observed protective effect of polyinosinic-polycytidylic acid on liver fibrosis was diminished through either depletion of natural killer cells or by disruption of the interferon gamma gene. Expression of retinoic acid early inducible 1, the NKG2D ligand, was undetectable on quiescent hepatic stellate cells, whereas high levels were found on activated hepatic stellate cells, which correlated with the resistance and susceptibility of quiescent hepatic stellate cells and activated hepatic stellate cells to natural killer cell lysis, respectively. Moreover, treatment with polyinosinic-polycytidylic acid or interferon gamma enhanced the cytotoxicity of natural killer cells against activated hepatic stellate cells and increased the expression of NKG2D and tumor necrosis factor-related apoptosis-inducing ligand on liver natural killer cells. Blocking NKG2D or tumor necrosis factor-related apoptosis-inducing ligand with neutralizing antibodies markedly diminished the cytotoxicity of polyinosinic-polycytidylic acid-activated natural killer cells against activated hepatic stellate cells. CONCLUSIONS: Our findings suggest that natural killer cells kill activated hepatic stellate cells via retinoic acid early inducible 1/NKG2D-dependent and tumor necrosis factor-related apoptosis-inducing ligand-dependent mechanisms, thereby ameliorating liver fibrosis.


Subject(s)
Apoptosis/immunology , Killer Cells, Natural/immunology , Liver Cirrhosis, Experimental/immunology , Receptors, Immunologic/physiology , Animals , DNA Primers , Disease Models, Animal , Flow Cytometry , Hepatitis, Viral, Animal/immunology , Ligands , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K , Perforin , Pore Forming Cytotoxic Proteins , Receptors, Immunologic/genetics , Receptors, Natural Killer Cell , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/physiology
4.
Biochem Biophys Res Commun ; 338(4): 1943-9, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16288983

ABSTRACT

The precise role of IL-6 in liver regeneration and hepatocyte proliferation is controversial and the role of SOCS3 in liver regeneration remains unknown. Here we show that in vitro treatment with IL-6 inhibited primary mouse hepatocyte proliferation. IL-6 induced p21cip1 protein expression in primary mouse hepatocytes. Disruption of the p21cip1 gene abolished the inhibitory effect of IL-6 on cell proliferation. Co-culture with nonparenchymal liver cells diminished IL-6 inhibition of hepatocyte proliferation, which was likely due to IL-6 stimulation of nonparenchymal cells to produce HGF. Finally, IL-6 induced higher levels of p21cip1 protein expression and a slightly stronger inhibition of cell proliferation in SOCS3+/- mouse hepatocytes compared to wild-type hepatocytes, while liver regeneration was enhanced and prolonged in SOCS3+/- mice. Our findings suggest that IL-6 directly inhibits hepatocyte proliferation via a p21cip1-dependent mechanism and indirectly enhances hepatocyte proliferation via stimulating nonparenchymal cells to produce HGF. SOCS3 negatively regulates liver regeneration.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Hepatocyte Growth Factor/biosynthesis , Hepatocytes/cytology , Interleukin-6/physiology , Liver Regeneration/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Animals , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/physiology , Hepatectomy , Hepatocytes/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Recombinant Proteins , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein
5.
Cell Mol Immunol ; 2(1): 40-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-16212910

ABSTRACT

It is well-documented that alcohol drinking together with hepatitis viral infection accelerates liver injury; however the underlying mechanisms remain unknown. In this paper, we demonstrated that primary hepatocytes from transgenic mice overexpressing hepatitis B virus X protein (HBX) were more susceptible to ethanol- and TNF-alpha-induced apoptotic killing. Compared to normal control mouse hepatocytes, ethanol and/or TNF-alpha treatment led to a significant increase in reactive oxygen species, mitochondrial permeability transition, cytochrome C release, caspase-3 activity, and poly (ADP-ribose) polymerase degradation in hepatocytes from HBX transgenic mice. Blocking caspase-3 activity antagonized ethanol- and TNF-alpha-induced apoptosis in primary hepatocytes from HBX transgenic mice. Taken together, our findings suggest that HBX sensitizes primary mouse hepatocytes to ethanol- and TNF-alpha-induced apoptosis by a caspase-3-dependent mechanism, which may partly explain the synergistic effects of alcohol consumption and hepatitis B virus infection on liver injury.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Ethanol/pharmacology , Hepatocytes/cytology , Hepatocytes/drug effects , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Caspase 3 , Cell Membrane Permeability/drug effects , Cells, Cultured , Hepatocytes/metabolism , Male , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins
6.
Am J Physiol Gastrointest Liver Physiol ; 287(5): G1044-52, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15246962

ABSTRACT

We have previously shown that IFN-gamma/STAT1 plays an essential role in concanavalin A (ConA)-induced T cell hepatitis via activation of apoptotic signaling pathways. Here we demonstrate that IFN-gamma/STAT1 also plays a crucial role in leukocyte infiltration into the liver in T cell hepatitis. After injection of ConA, leukocytes were significantly infiltrated into the liver, which was suppressed in IFN-gamma(-/-) and STAT1(-/-) mice. Disruption of the IFN regulatory factor-1 (IRF-1) gene, a downstream target of IFN-gamma/STAT1, abolished ConA-induced liver injury and suppressed leukocyte infiltration into the liver. Additionally, ConA injection induced expression of a wide variety of chemokines and adhesion molecules in the liver. Among them, expression of ICAM-1, VCAM-1, monokine induced by IFN-gamma (Mig), CC chemokine ligand-20, epithelial cell-derived neutrophil-activating peptide (ENA)-78, IFN-inducible T cell-alpha chemoattractant (I-TAC), and IFN-inducible protein-10 (IP-10) was markedly attenuated in IFN-gamma(-/-), STAT1(-/-), and IRF-1(-/-) mice. In primary mouse hepatocytes, Kupffer cells, and endothelial cells, in vitro treatment with IFN-gamma activated STAT1, STAT3, and IRF-1, and induced expression of VCAM-1, ICAM-1, Mig, ENA-78, I-TAC, and IP-10 mRNA. Induction of these chemokines and adhesion molecules was markedly diminished in STAT1(-/-) and IRF-1(-/-) hepatic cells compared with wild-type hepatic cells. These findings suggest that in addition to induction of apoptosis, previously well documented, IFN-gamma also stimulated hepatocytes, sinusoidal endothelial cells, and Kupffer cells partly via an STAT1/IRF-1-dependent mechanism to produce multiple chemokines and adhesive molecules responsible for promoting infiltration of leukocytes and, ultimately, resulting in hepatitis.


Subject(s)
DNA-Binding Proteins/metabolism , Hepatitis, Animal/immunology , Inflammation Mediators , Interferon-gamma/metabolism , Phosphoproteins/metabolism , Signal Transduction , T-Lymphocytes/immunology , Trans-Activators/metabolism , Animals , Cell Adhesion Molecules/biosynthesis , Chemokines/biosynthesis , Concanavalin A , Endothelial Cells/metabolism , Eosinophils , Hepatitis, Animal/chemically induced , Hepatitis, Animal/metabolism , Hepatitis, Animal/pathology , Hepatocytes/metabolism , Interferon Regulatory Factor-1 , Interferon-gamma/pharmacology , Kupffer Cells/metabolism , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , STAT1 Transcription Factor
7.
Am J Physiol Gastrointest Liver Physiol ; 287(2): G471-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15064234

ABSTRACT

Alcohol consumption is a major risk factor accelerating the progression of liver disease in patients with chronic hepatitis virus infection. However, the mechanism underlying the enhanced susceptibility of alcoholics to liver injury is not fully understood. Here, we demonstrate that chronic ethanol consumption increases the susceptibility of C57BL/6 mice to concanavalin A (Con A)-induced T cell-mediated hepatitis. Injection of a low dose of Con A (5 microg/g) causes severe liver damage in ethanol-fed mice as evidenced by a significant elevation of serum alanine aminotransaminase levels, massive necrosis, and infiltration of leukocytes but only slightly induces liver injury in control pair-fed mice. In ethanol-fed mice, the activation and cytotoxicity of natural killer T cells, cells that play key roles in Con A-induced T cell hepatitis, are not significantly enhanced relative to pair-fed mice. Moreover, Con A-induced activation of hepatic NF-kappaB is increased, whereas activation of STAT1 and STAT3 is attenuated in ethanol-fed mice. Consistent with this result, the expression of chemokines and adhesion molecules [such as ICAM-1, macrophage inflammatory protein (MIP)-1, MIP-2, and MCP-1] controlled by NF-kappaB is upregulated, whereas STAT1-controlled expression of chemokines (such as MIG and IP-10) is downregulated in ethanol-fed mice compared with pair-fed mice. In conclusion, chronic alcohol consumption accelerates T cell-mediated hepatitis via upregulation of the NF-kappaB signaling pathway and subsequently enhances expression of chemokines/adhesive molecules and recruitment of leukocytes into the liver. Downregulation of the antiapoptotic STAT3 signal may also contribute to alcohol potentiation of T cell hepatitis.


Subject(s)
Alcohol Drinking , Chemical and Drug Induced Liver Injury/pathology , DNA-Binding Proteins/metabolism , Ethanol/pharmacology , Liver/pathology , NF-kappa B/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Animals , Cell Adhesion Molecules/metabolism , Chemokines/metabolism , Concanavalin A/adverse effects , Hepatocytes , Killer Cells, Natural/drug effects , Leukocytes/pathology , Liver/physiopathology , Male , Mice , Mice, Inbred C57BL , Monocytes , STAT3 Transcription Factor , T-Lymphocytes , Time Factors
8.
Biochem J ; 379(Pt 1): 199-208, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14690454

ABSTRACT

IFN-gamma (interferon-gamma) modulates IFN-alpha therapy in chronic hepatitis C infection; however, the underlying mechanism remains unclear. Here we demonstrate that long-term (3-6 days) but not short-term (up to 1 day) IFN-gamma treatment of human hepatoma Hep3B cells attenuates IFN-alpha activation of STAT1 (signal transducers and activators of transcription factor 1), STAT2 and STAT3, but enhances IFN-gamma and interleukin 6 activation of STATs. Prolonged exposure to IFN-gamma also significantly induces STAT1 protein expression without affecting STAT2, STAT3 and ERK (extracellular-signal-regulated kinase) 1/2 protein expression. To determine the role of STAT1 protein overexpression in regulation of IFN-alpha signalling, Hep3B cells were stably transfected with wild-type STAT1. Overexpression of STAT1 via stable transfection enhances IFN-gamma activation of STAT1, but surprisingly attenuates IFN-alpha activation of STAT1, STAT2 and STAT3 without affecting Janus kinase activation. This STAT1-mediated inhibition does not require STAT1 tyrosine phosphorylation because overexpression of dominant-negative STAT1 with a mutation on tyrosine residue 701 also blocks IFN-alpha activation of STAT1, STAT2 and STAT3. Moreover, overexpression of STAT1 blocks IFN-alpha-activated STAT2 translocation from IFN-alpha receptor 2 to IFN-alpha receptor 1, a critical step in IFN-alpha signalling activation. Finally, significantly higher levels of STAT1 protein expression, which is probably induced by IFN-gamma, are detected in the majority of hepatitis C virus-infected livers compared with healthy controls. In conclusion, long-term IFN-gamma treatment inhibits IFN-alpha-activated signals most probably, at least in part, through the induction of STAT1 protein expression, which could partly contribute to IFN-alpha treatment failure in hepatitis C patients.


Subject(s)
Antiviral Agents/pharmacology , DNA-Binding Proteins/physiology , Gene Expression Regulation/drug effects , Hepatitis C, Chronic/drug therapy , Interferon-alpha/antagonists & inhibitors , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins , Liver/metabolism , Trans-Activators/physiology , Carcinoma, Hepatocellular/pathology , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Cycle , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Genes, Reporter , Hepatitis C, Chronic/metabolism , Humans , Interferon-alpha/physiology , Interleukin-6/metabolism , Janus Kinase 1 , Liver/drug effects , Liver Cirrhosis/metabolism , Liver Neoplasms/pathology , Membrane Proteins , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/biosynthesis , Receptor, Interferon alpha-beta , Receptors, Interferon/metabolism , Recombinant Fusion Proteins/physiology , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , STAT1 Transcription Factor , STAT2 Transcription Factor , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins , TYK2 Kinase , Trans-Activators/biosynthesis , Trans-Activators/genetics , Trans-Activators/metabolism , Transfection
9.
J Immunol ; 171(6): 3233-44, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960353

ABSTRACT

T cell-mediated immune responses are implicated in the pathogenesis of a variety of liver disorders; however, the underlying mechanism remains obscure. Con A injection is a widely accepted mouse model to study T cell-mediated liver injury, in which STAT6 is rapidly activated. Disruption of the IL-4 and STAT6 gene by way of genetic knockout abolishes Con A-mediated liver injury without affecting IFN-gamma/STAT1, IL-6/STAT3, or TNF-alpha/NF-kappaB signaling or affecting NKT cell activation. Infiltration of neutrophils and eosinophils in Con A-induced hepatitis is markedly suppressed in IL-4 (-/-) and STAT6(-/-) mice compared with wild-type mice. IL-4 treatment induces expression of eotaxins in hepatocytes and sinusoidal endothelial cells isolated from wild-type mice but not from STAT6(-/-) mice. Con A injection induces expression of eotaxins in the liver and elevates serum levels of IL-5 and eotaxins; such induction is markedly attenuated in IL-4(-/-) and STAT6(-/-) mice. Finally, eotaxin blockade attenuates Con A-induced liver injury and leukocyte infiltration. Taken together, these findings suggest that IL-4/STAT6 plays a critical role in Con A-induced hepatitis, via enhancing expression of eotaxins in hepatocytes and sinusoidal endothelial cells, and induces IL-5 expression, thereby facilitating recruitment of eosinophils and neutrophils into the liver and resulting in hepatitis.


Subject(s)
Cell Movement/immunology , Chemokines, CC/biosynthesis , Hepatitis, Animal/immunology , Interleukin-4/physiology , Interleukin-5/biosynthesis , Leukocytes/pathology , T-Lymphocyte Subsets/immunology , Trans-Activators/physiology , Adoptive Transfer , Animals , Cell Movement/genetics , Cells, Cultured , Chemokine CCL11 , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/immunology , Concanavalin A/administration & dosage , DNA-Binding Proteins/physiology , Eosinophils/pathology , Hepatitis, Animal/chemically induced , Hepatitis, Animal/genetics , Hepatitis, Animal/pathology , Immune Sera/administration & dosage , Injections, Intravenous , Interferon-gamma/physiology , Interleukin-4/deficiency , Interleukin-4/genetics , Killer Cells, Natural/immunology , Leukocytes, Mononuclear/transplantation , Liver/immunology , Liver/metabolism , Liver/pathology , Lymphocyte Activation/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/physiology , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , STAT1 Transcription Factor , STAT3 Transcription Factor , STAT6 Transcription Factor , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/immunology , Spleen/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/genetics , Up-Regulation/immunology
10.
Gastroenterology ; 125(1): 202-15, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12851884

ABSTRACT

BACKGROUND & AIMS: Orthotopic liver transplantation is currently the only curative therapy for chronic end-stage liver disease and acute liver failure. However, a scarcity of cadaveric donors has led to a critical shortage of organs available for transplant. This is further complicated by the prevalence of steatosis in about 13%-50% of donor livers, which is associated with a high risk of dysfunction and primary nonfunction. METHODS: Steatotic Zucker rat livers and livers from alcohol-fed rats were transplanted into lean control rats. Liver injury, activation of survival signals, and hepatic microcirculation were compared in nontreated and interleukin-6 (IL-6)-treated steatotic isografts. RESULTS: IL-6 pretreatment of steatotic Zucker rat liver isografts dramatically reduces mortality and liver injury following transplantation. Reperfusion after transplantation induces significant sinusoidal endothelial cell necrapoptosis in steatotic Zucker rat liver isografts, which is prevented by in vitro IL-6 pretreatment. IL-6 treatment activates cell survival signal transducer and activator of transcription factor 3 (STAT3) in hepatocytes and sinusoidal endothelial cells. Laser Doppler imaging and microsphere analyses demonstrate that IL-6 treatment markedly improves hepatic microcirculation, which is impaired in steatotic Zucker rat liver transplants. Finally, in vitro IL-6 treatment of donor livers also markedly reduces mortality associated with fatty liver transplants from alcohol-fed rats. CONCLUSIONS: IL-6 induces hepatoprotection of steatotic liver isografts via preventing sinusoidal endothelial cell necrapoptosis and consequent amelioration of hepatic microcirculation, and protecting against hepatocyte death. IL-6 pretreatment of steatotic livers may render such allografts useable for clinical transplantation.


Subject(s)
Fatty Liver, Alcoholic/drug therapy , Fatty Liver, Alcoholic/surgery , Interleukin-6/pharmacology , Liver Transplantation/mortality , Animals , Central Nervous System Depressants/pharmacology , Combined Modality Therapy , DNA-Binding Proteins/metabolism , Disease Models, Animal , Endothelium/cytology , Ethanol/pharmacology , Fatty Liver, Alcoholic/mortality , Graft Survival/drug effects , In Vitro Techniques , Liver/blood supply , Liver/cytology , Liver/drug effects , Liver Circulation/drug effects , Liver Failure/drug therapy , Liver Failure/mortality , Liver Failure/surgery , Microcirculation , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Zucker , STAT3 Transcription Factor , Survival Rate , Trans-Activators/metabolism , bcl-X Protein
11.
Am J Physiol Gastrointest Liver Physiol ; 285(4): G761-8, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12816762

ABSTRACT

Interferon-gamma (IFN-gamma) has been implicated in liver damage in animal models and chronic hepatitis C infection; however, the underlying mechanism is not clear. Here we examined the role of STAT1, a key signaling molecule for IFN-gamma, in a model of murine hepatitis induced by the injection of LPS/D-galactosamine and in human hepatoma Hep3B cells. STAT1 is rapidly activated and highly induced after injection of LPS/D-galactosamine. Both overexpression of STAT1 and hepatocellular damage are located in the same pericentral region. Disruption of the STAT1 gene abolishes LPS/D-galactosamine-induced liver injury. Studies from IFN-gamma-deficient mice indicate that IFN-gamma is the major cytokine responsible for activation and hyperexpression of STAT1 in LPS/D-galactosamine-induced hepatitis. Hep3B cells overexpressing dominant negative STAT1 are resistant to IFN-gamma and IFN-gamma + TNF-alpha-induced cell death, whereas Hep3B cells overexpressing wild-type STAT1 are more susceptible to cell death. Taken together, these findings suggest that STAT1 plays an essential role in LPS/D-galactosamine-induced liver apoptosis and injury.


Subject(s)
Apoptosis/drug effects , Chemical and Drug Induced Liver Injury , DNA-Binding Proteins/physiology , Galactosamine/pharmacology , Lipopolysaccharides/pharmacology , Trans-Activators/physiology , Animals , Cell Line , Chemical and Drug Induced Liver Injury/etiology , DNA Fragmentation , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Flow Cytometry , Hepatocytes/chemistry , Hepatocytes/ultrastructure , Interferon-gamma/deficiency , Interferon-gamma/pharmacology , Interferon-gamma/physiology , Liver Diseases/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Phosphorylation , Phosphotyrosine/metabolism , STAT1 Transcription Factor , Trans-Activators/deficiency , Trans-Activators/genetics , Transfection , Tumor Necrosis Factor-alpha/pharmacology
12.
Pol Merkur Lekarski ; 13(74): 126-8, 2002 Aug.
Article in Polish | MEDLINE | ID: mdl-12420343

ABSTRACT

UNLABELLED: The aim of the study was evaluation of serum neopterin and beta 2-microglobulin concentrations as "prognostic markers" of AIDS natural history. MATERIAL AND METHODS: Investigations was performed in 68 patients (48 hiv-infected and 20 without hiv-infection), who were divided into 3 groups according to HIV CDC classification. RESULTS: The highest serum concentrations of neopterin and beta 2-microglobulin were detected in patients with AIDS. In a non-symptomatic HIV-infected patients p24 Ag was absent. It was detected in patients with AIDS. Negative correlation between neopterin and beta 2-microglobulin serum concentration and CD4 count was detected in sera from all patients. Positive correlation was observed between p24 Ag, serum neopterin and beta 2-microglobulin concentration in patients with AIDS. CONCLUSIONS: Progression of HIV infection was connected with an increase of serum neopterin and beta 2-microglobulin serum concentrations. Neopterin serum concentration seems to be a more sensitive marker of HIV infection than beta 2-microglobulin.


Subject(s)
Acquired Immunodeficiency Syndrome/blood , Neopterin/blood , beta 2-Microglobulin/blood , Acquired Immunodeficiency Syndrome/immunology , Adult , Biomarkers/blood , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Disease Progression , Female , HIV Core Protein p24/blood , Humans , Male , Prognosis
13.
J Clin Invest ; 110(10): 1503-13, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12438448

ABSTRACT

T cell-mediated fulminant hepatitis is a life-threatening event for which the underlying mechanism is not fully understood. Injection of concanavalin A (Con A) into mice recapitulates the histological and pathological sequelae of T cell-mediated hepatitis. In this model, both signal transducer and activator of transcription factor 1 (STAT1) and STAT3 are activated in the liver. Disruption of the STAT1 gene by way of genetic knockout attenuates liver injury, suppresses CD4(+) and NK T cell activation, and downregulates expression of proapoptotic interferon regulatory factor-1 protein and suppressor of cytokine signaling-1 (SOCS1), but enhances STAT3 activation and STAT3-controlled antiapoptotic signals. Studies from IFN-gamma-deficient mice indicate that IFN-gamma not only is the major cytokine responsible for STAT1 activation but also partially accounts for STAT3 activation. Moreover, downregulation of STAT3 activation in IL-6-deficient mice is associated with decreased STAT3-controlled antiapoptotic signals and expression of SOCS3, but upregulation of STAT1 activation and STAT1-induced proapoptotic signals and exacerbation of liver injury. Taken together, these findings suggest that STAT1 plays a harmful role in Con A-mediated hepatitis by activation of CD4(+) and NK T cells and directly inducing hepatocyte death, whereas STAT3 protects against liver injury by suppression of IFN-gamma signaling and induction of antiapoptotic protein Bcl-X(L). STAT1 and STAT3 in hepatocytes also negatively regulate one another through the induction of SOCS.


Subject(s)
DNA-Binding Proteins/metabolism , Hepatitis, Autoimmune/metabolism , Intracellular Signaling Peptides and Proteins , Proteins/metabolism , Repressor Proteins , T-Lymphocytes/immunology , Trans-Activators/metabolism , Transcription Factors , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Concanavalin A/toxicity , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Hepatitis, Autoimmune/etiology , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interleukin-6/deficiency , Interleukin-6/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT1 Transcription Factor , STAT3 Transcription Factor , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Trans-Activators/deficiency , Trans-Activators/genetics
14.
Gastroenterology ; 122(4): 1020-34, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11910354

ABSTRACT

BACKGROUND & AIMS: Interferon (IFN)-alpha therapy is currently the primary choice for viral hepatitis and a promising treatment for hepatocellular carcinoma (HCC). Primary mouse and rat hepatocytes respond poorly to IFN-alpha stimulation. Thus, it is very important to examine the IFN-alpha signal pathway in primary human hepatocytes. METHODS: The IFN-alpha-activated signals and genes in primary human hepatocytes and hepatoma cells were examined by Western blotting and microarray analyses. RESULTS: Primary human hepatocytes respond very well to IFN-alpha stimulation as shown by activation of multiple signal transducer and activator of transcription factor (STAT) 1, 2, 3, 5, and multiple genes. The differential response to IFN-alpha stimulation in primary human and mouse hepatocytes may be caused by expression of predominant functional IFN-alpha receptor 2c (IFNAR2c) in primary human hepatocytes vs. expression of predominant inhibitory IFNAR2a in mouse hepatocytes. Microarray analyses of primary human hepatocytes show that IFN-alpha up-regulates about 44 genes by over 2-fold and down-regulates about 9 genes by 50%. The up-regulated genes include a variety of antiviral and tumor suppressors/proapoptotic genes. The down-regulated genes include c-myc and c-Met, the hepatocyte growth factor (HGF) receptor. Down-regulation of c-Met is caused by IFN-alpha suppression of the c-Met promoter through down-regulation of Sp1 binding and results in attenuation of HGF-induced signals and cell proliferation. CONCLUSIONS: IFN-alpha directly targets human hepatocytes, followed by activation of multiple STATs and regulation of a wide variety of genes, which may contribute to the antiviral and antitumor activities of IFN-alpha in human liver.


Subject(s)
DNA-Binding Proteins/metabolism , Hepatocytes/metabolism , Immunologic Factors/pharmacology , Interferon-alpha/pharmacology , Milk Proteins , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/drug effects , Trans-Activators/metabolism , Animals , Antiviral Agents/pharmacology , Carcinoma, Hepatocellular , Cell Division/drug effects , Down-Regulation/drug effects , Female , Gene Expression/drug effects , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Interferon-gamma/pharmacology , Liver Neoplasms , Male , Membrane Proteins , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-met/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , STAT1 Transcription Factor , STAT2 Transcription Factor , STAT3 Transcription Factor , STAT5 Transcription Factor , Solubility , Sp1 Transcription Factor/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
15.
Free Radic Biol Med ; 32(5): 414-20, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11864781

ABSTRACT

There is evidence suggesting that patients infected with human immunodeficiency virus (HIV) are under chronic oxidative stress. In the present study, the level of oxidatively modified bases in lymphocyte DNA and some other parameters of oxidative stress were measured in HIV-infected patients (n = 30), as well as in control groups (10 healthy volunteers and 15 HIV-seronegative injected drug users). Additional experiments were conducted using lymphocyte DNA samples from asymptomatic seropositive, HIV-infected patients who were supplemented with antioxidant vitamins A, C, and E or received placebo. Significant increases in the amount of the modified DNA bases were observed in HIV-infected patients when compared with the control group. The concentration of thiobarbituric acid reactive substances (TBARS) was higher and activities of antioxidant enzymes (superoxide dismutase and catalase) were lower in the group of HIV-infected patients in comparison to the control group. Vitamin supplementation resulted in the significant decrease in the levels of all modified DNA bases when compared to the patients who received placebo. The reduction of TBARS and the restoration of the activity of the enzymes were also observed. Our data suggest that people infected with HIV can benefit from treatment with antioxidant vitamins.


Subject(s)
Antioxidants/pharmacology , Ascorbic Acid/pharmacology , DNA/metabolism , HIV Infections/blood , Lymphocytes/drug effects , Vitamin A/pharmacology , Vitamin E/pharmacology , Adolescent , Adult , Antiretroviral Therapy, Highly Active , Case-Control Studies , Catalase/metabolism , Chromatography, High Pressure Liquid , DNA Damage/drug effects , DNA Repair , Dietary Supplements , Female , HIV Infections/drug therapy , Humans , Lipid Peroxidation , Lymphocytes/metabolism , Lymphocytes/virology , Male , Mass Spectrometry , Oxidation-Reduction , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances
16.
Oncogene ; 21(1): 32-43, 2002 Jan 03.
Article in English | MEDLINE | ID: mdl-11791174

ABSTRACT

Elevation of serum interleukin-6 (IL-6) levels is always associated with alcoholic liver disease (ALD), but the significance of such elevation is not clear. Here we show that chronic ethanol consumption induces significant apoptosis in the liver of IL-6 (-/-) mice but not IL-6 (+/+) mice. IL-6 (-/-) hepatocytes are more susceptible to ethanol- and tumor necrosis factor alpha- (TNFalpha-) induced apoptotic killing, which can be corrected by IL-6. Expression of both anti-apoptotic (such as Bcl-2 and Bcl-x(L)) and proapoptotic (such as Bax) proteins is markedly elevated in the liver of human ALD and chronically ethanol-fed IL-6 (+/+) mice. On the contrary, induction of Bcl-2 and Bcl-x(L) is not observed in the liver of chronically ethanol-fed IL-6 (-/-) mice, whereas expression of Bax protein remains elevated. Injection of IL-6 markedly induces expression of Bcl-2 and Bcl-x(L) but not Bax in the liver. Finally, high concentrations of ethanol inhibit IL-6-activated anti-apoptotic signal, but increasing the concentrations of IL-6 is able to overcome such inhibitory effect. These findings suggest that elevated serum IL-6 levels in ALD may overcome the inhibitory effect of ethanol on IL-6-mediated anti-apoptotic signals and prevent alcohol-induced hepatic apoptosis by induction of Bcl-2 and Bcl-x(L).


Subject(s)
Alcoholism/metabolism , Apoptosis/drug effects , Ethanol/toxicity , Interleukin-6/physiology , Liver Diseases, Alcoholic/genetics , Liver/drug effects , Proto-Oncogene Proteins c-bcl-2/physiology , Animals , Body Weight/drug effects , Cells, Cultured/drug effects , Gene Expression Regulation/drug effects , Genes, bcl-2 , Genetic Predisposition to Disease , Genotype , Hepatocytes/drug effects , Humans , Interleukin-6/blood , Interleukin-6/genetics , Liver/pathology , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Knockout , Organ Size/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Tumor Necrosis Factor-alpha/toxicity , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...