Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Gastroenterology ; 144(5): 1042-1054.e4, 2013 May.
Article in English | MEDLINE | ID: mdl-23391818

ABSTRACT

BACKGROUND & AIMS: Transforming growth factor (TGF)-Ɵ-activated kinase 1 (TAK1) is activated in different cytokine signaling pathways. Deletion of Tak1 from hepatocytes results in spontaneous development of hepatocellular carcinoma (HCC), liver inflammation, and fibrosis. TGF-Ɵ activates TAK1 and Smad signaling, which regulate cell death, proliferation, and carcinogenesis. However, it is not clear whether TGF-Ɵ signaling in hepatocytes, via TGF-Ɵ receptor-2 (Tgfbr2), promotes HCC and liver fibrosis. METHODS: We generated mice with hepatocyte-specific deletion of Tak1 (Tak1ΔHep), as well as Tak1/Tgfbr2DHep and Tak1/Smad4ΔHep mice. Tak1flox/flox, Tgfbr2ΔHep, and Smad4ΔHep mice were used as controls, respectively. We assessed development of liver injury, inflammation, fibrosis, and HCC. Primary hepatocytes isolated from these mice were used to assess TGF-Ɵ-mediated signaling. RESULTS: Levels of TGF-Ɵ, TGF-ƟR2, and phospho-Smad2/3 were increased in HCCs from Tak1ΔHep mice, which developed liver fibrosis and inflammation by 1 month and HCC by 9 months. However, Tak1/Tgfbr2ΔHep mice did not have this phenotype, and their hepatocytes did not undergo spontaneous cell death or compensatory proliferation. Hepatocytes from Tak1ΔHep mice incubated with TGF-Ɵ did not activate p38, c-Jun N-terminal kinase, or nuclear factor-κB; conversely, TGF-Ɵ-mediated cell death and phosphorylation of Smad2/3 were increased, compared with control hepatocytes. Blocking the Smad pathway inhibited TGF-Ɵ-mediated death of Tak1-/- hepatocytes. Accordingly, disruption of Smad4 reduced the spontaneous liver injury, inflammation, fibrosis, and HCC that develops in Tak1ΔHep mice. Levels of the anti-apoptotic protein Bcl-xL, Ɵ-catenin, connective tissue growth factor, and vascular endothelial growth factor were increased in HCC from Tak1ΔHep mice, but not in HCCs from Tak1/Tgfbr2ΔHep mice. Injection of N-nitrosodiethylamine induced HCC formation in wild-type mice, but less in Tgfbr2ΔHep mice. CONCLUSIONS: TGF-Ɵ promotes development of HCC in Tak1ΔHep mice by inducing hepatocyte apoptosis and compensatory proliferation during early phases of tumorigenesis, and inducing expression of anti-apoptotic, pro-oncogenic, and angiogenic factors during tumor progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA, Neoplasm/genetics , Gene Deletion , Liver Cirrhosis/pathology , Liver Neoplasms, Experimental/genetics , MAP Kinase Kinase Kinases/genetics , Transforming Growth Factor beta/genetics , Animals , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , DNA Mutational Analysis , Hepatocytes/metabolism , Hepatocytes/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Transgenic , Phenotype , Signal Transduction/genetics , Transforming Growth Factor beta/metabolism , Tumor Cells, Cultured
2.
Proc Natl Acad Sci U S A ; 107(2): 844-9, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20080763

ABSTRACT

TGF-beta-activated kinase 1 (TAK1) is a MAP3K family member that activates NF-kappaB and JNK via Toll-like receptors and the receptors for IL-1, TNF-alpha, and TGF-beta. Because the TAK1 downstream molecules NF-kappaB and JNK have opposite effects on cell death and carcinogenesis, the role of TAK1 in the liver is unpredictable. To address this issue, we generated hepatocyte-specific Tak1-deficient (Tak1DeltaHEP) mice. The Tak1DeltaHEP mice displayed spontaneous hepatocyte death, compensatory proliferation, inflammatory cell infiltration, and perisinusoidal fibrosis at age 1 month. Older Tak1DeltaHEP mice developed multiple cancer nodules characterized by increased expression of fetal liver genes including alpha-fetoprotein. Cultures of primary hepatocytes deficient in Tak1 exhibited spontaneous cell death that was further increased in response to TNF-alpha. TNF-alpha increased caspase-3 activity but activated neither NF-kappaB nor JNK in Tak1-deficient hepatocytes. Genetic abrogation of TNF receptor type I (TNFRI) in Tak1DeltaHEP mice reduced liver damage, inflammation, and fibrosis compared with unmodified Tak1DeltaHEP mice. In conclusion, hepatocyte-specific deletion of TAK1 in mice resulted in spontaneous hepatocyte death, inflammation, fibrosis, and carcinogenesis that was partially mediated by TNFR signaling, indicating that TAK1 is an essential component for cellular homeostasis in the liver.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Inflammation/pathology , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Liver/pathology , Adaptor Proteins, Signal Transducing/deficiency , Alanine Transaminase/blood , Animals , Apoptosis , Cell Division/physiology , Female , Gene Deletion , Hepatocytes/pathology , Hepatocytes/physiology , Liver Regeneration/genetics , Male , Mice , Sex Characteristics
3.
Hepatology ; 53(5): 1730-41, 2011 May.
Article in English | MEDLINE | ID: mdl-21384410

ABSTRACT

UNLABELLED: Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a multicomponent enzyme that mediates electron transfer from nicotinamide adenine dinucleotide phosphate to molecular oxygen, which leads to the production of superoxide. NOX2/gp91(phox) is a catalytic subunit of NOX expressed in phagocytic cells. Several homologues of NOX2, including NOX1, have been identified in nonphagocytic cells. We investigated the contributory role of NOX1 and NOX2 in hepatic fibrosis. Hepatic fibrosis was induced in wild-type (WT) mice, NOX1 knockout (NOX1KO) mice, and NOX2 knockout (NOX2KO) mice by way of either carbon tetrachloride (CCl(4) ) injection or bile duct ligation (BDL). The functional contribution of NOX1 and NOX2 in endogenous liver cells, including hepatic stellate cells (HSCs), and bone marrow (BM)-derived cells, including Kupffer cells (KCs), to hepatic reactive oxygen species (ROS) generation and hepatic fibrosis was assessed in vitro and in vivo using NOX1 or NOX2 BM chimeric mice. Hepatic NOX1 and NOX2 messenger RNA expression was increased in the two experimental mouse models of hepatic fibrosis. Whereas NOX1 was expressed in HSCs but not in KCs, NOX2 was expressed in both HSCs and KCs. Hepatic fibrosis and ROS generation were attenuated in both NOX1KO and NOX2KO mice after CCl(4) or BDL. Liver fibrosis in chimeric mice indicated that NOX1 mediates the profibrogenic effects in endogenous liver cells, whereas NOX2 mediates the profibrogenic effects in both endogenous liver cells and BM-derived cells. Multiple NOX1 and NOX2 components were up-regulated in activated HSCs. Both NOX1- and NOX2-deficient HSCs had decreased ROS generation and failed to up-regulate collagen α1(I) and transforming growth factor Ɵ in response to angiotensin II. CONCLUSION: Both NOX1 and NOX2 have an important role in hepatic fibrosis in endogenous liver cells, including HSCs, whereas NOX2 has a lesser role in BM-derived cells.


Subject(s)
Liver Cirrhosis/etiology , Membrane Glycoproteins/physiology , NADH, NADPH Oxidoreductases/physiology , NADPH Oxidases/physiology , Animals , Bile Ducts , Carbon Tetrachloride/pharmacology , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/physiology , Ligation , Liver/cytology , Liver/drug effects , Liver/metabolism , Mice , Mice, Knockout , NADPH Oxidase 1 , NADPH Oxidase 2 , Reactive Oxygen Species
4.
Gastroenterology ; 139(1): 323-34.e7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20347818

ABSTRACT

BACKGROUND & AIMS: Development of nonalcoholic steatohepatitis (NASH) involves the innate immune system and is mediated by Kupffer cells and hepatic stellate cells (HSCs). Toll-like receptor 9 (TLR9) is a pattern recognition receptor that recognizes bacteria-derived cytosine phosphate guanine (CpG)-containing DNA and activates innate immunity. We investigated the role of TLR9 signaling and the inflammatory cytokine interleukin-1beta (IL-1beta) in steatohepatitis, fibrosis, and insulin resistance. METHODS: Wild-type (WT), TLR9(-/-), IL-1 receptor (IL-1R)(-/-), and MyD88(-/-) mice were fed a choline-deficient amino acid-defined (CDAA) diet for 22 weeks and then assessed for steatohepatitis, fibrosis, and insulin resistance. Lipid accumulation and cell death were assessed in isolated hepatocytes. Kupffer cells and HSCs were isolated to assess inflammatory and fibrogenic responses, respectively. RESULTS: The CDAA diet induced NASH in WT mice, characterized by steatosis, inflammation, fibrosis, and insulin resistance. TLR9(-/-) mice showed less steatohepatitis and liver fibrosis than WT mice. Among inflammatory cytokines, IL-1beta production was suppressed in TLR9(-/-) mice. Kupffer cells produced IL-1beta in response to CpG oligodeoxynucleotide. IL-1beta but not CpG-oligodeoxynucleotides, increased lipid accumulation in hepatocytes. Lipid accumulation in hepatocytes led to nuclear factor-kappaB inactivation, resulting in cell death in response to IL-1beta. IL-1beta induced fibrogenic responses in HSCs, including secretion of tissue inhibitor of metalloproteinase-1. IL-1R(-/-) mice had reduced steatohepatitis and fibrosis, compared with WT mice. Mice deficient in MyD88, an adaptor molecule for TLR9 and IL-1R signaling, also had reduced steatohepatitis and fibrosis. TLR9(-/-), IL-1R(-/-), and MyD88(-/-) mice had less insulin resistance than WT mice on the CDAA diet. CONCLUSIONS: In a mouse model of NASH, TLR9 signaling induces production of IL-1beta by Kupffer cells, leading to steatosis, inflammation, and fibrosis.


Subject(s)
Fatty Liver/etiology , Interleukin-1beta/physiology , Toll-Like Receptor 9/physiology , Animals , Choline Deficiency/complications , Hepatic Stellate Cells/physiology , Insulin Resistance , Interleukin-1beta/genetics , Kupffer Cells/physiology , Lipid Metabolism , Liver Cirrhosis, Experimental/etiology , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/physiology , RNA, Messenger/analysis , Signal Transduction
5.
Hepatology ; 52(4): 1390-400, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20683935

ABSTRACT

UNLABELLED: Chronic liver disease is associated with hepatocyte injury, inflammation, and fibrosis. Chemokines and chemokine receptors are key factors for the migration of inflammatory cells such as macrophages and noninflammatory cells such as hepatic stellate cells (HSCs). The expression of CX3CR1 and its ligand, CX3CL1, is up-regulated in chronic liver diseases such as chronic hepatitis C. However, the precise role of CX3CR1 in the liver is still unclear. Here we investigated the role of the CX3CL1-CX3CR1 interaction in a carbon tetrachloride (CCl(4))-induced liver inflammation and fibrosis model. CX3CR1 was dominantly expressed in Kupffer cells in the liver. In contrast, the main source of CX3CL1 was HSCs. Mice deficient in CX3CR1 showed significant increases in inflammatory cell recruitment and cytokine production [including tumor necrosis factor α (TNF-α); monocyte chemoattractant protein 1; macrophage inflammatory protein 1Ɵ; and regulated upon activation, normal T cell expressed, and secreted (RANTES)] after CCl(4) treatment versus wild-type (WT) mice. This suggested that CX3CR1 signaling prevented liver inflammation. Kupffer cells in CX3CR1-deficient mice after CCl(4) treatment showed increased expression of TNF-α and transforming growth factor Ɵ and reduced expression of the anti-inflammatory markers interleukin-10 (IL-10) and arginase-1. Coculture experiments showed that HSCs experienced significantly greater activation by Kupffer cells from CCl(4)-treated CX3CR1-deficient mice versus WT mice. Indeed, augmented fibrosis was observed in CX3CR1-deficient mice versus WT mice after CCl(4) treatment. Finally, CX3CL1 treatment induced the expression of IL-10 and arginase-1 in WT cultured Kupffer cells through CX3CR1, which in turn suppressed HSC activation. CONCLUSION: The CX3CL1-CX3CR1 interaction inhibits inflammatory properties in Kupffer cells/macrophages and results in decreased liver inflammation and fibrosis.


Subject(s)
Carbon Tetrachloride Poisoning/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Chemokine CX3CL1/physiology , Receptors, Chemokine/physiology , Animals , CX3C Chemokine Receptor 1 , Carbon Tetrachloride Poisoning/prevention & control , Cell Movement/drug effects , Coculture Techniques , Hepatic Stellate Cells/metabolism , Interleukin-10/biosynthesis , Kupffer Cells/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
6.
Alcohol Clin Exp Res ; 35(8): 1509-18, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21463341

ABSTRACT

BACKGROUND: Excessive alcohol intake causes an increase in intestinal permeability that induces translocation of gut-derived lipopolysaccharide (LPS) to the portal vein. Increased LPS in the portal vein stimulates Kupffer cells through Toll-like receptor (TLR) 4 in the liver. Activated TLR4 signaling in Kupffer cells induces various inflammatory mediators including TNF-α, IL-1Ɵ, and reactive oxygen species, resulting in liver injury. Hepatic stellate cells (HSCs) also express TLR4. This study investigates whether TLR4 on bone marrow (BM)-derived cells, including Kupffer cells, or non-BM-derived endogenous liver cells, including HSCs, contributes to the progression of alcohol-induced steatohepatitis and fibrogenesis in mice. METHODS: TLR4 BM chimera (wild-type [WT] mice with TLR4(-/-) BM or TLR4(-/-) mice with WT BM) were generated by the combination of liposomal clodronate injection with whole body irradiation and BM transplantation, followed by treatment with intragastric alcohol feeding. RESULTS: WT mice transplanted with WT BM exhibited liver injury, steatosis, inflammation, and a fibrogenic response. Conversely, TLR4(-/-) mice with TLR4(-/-) BM displayed less steatosis, liver injury, and inflammation. Notably, steatosis, macrophage infiltration, and alanine aminotransferase levels in both TLR4-chimeric mice showed intermediate levels between WT mice transplanted with WT BM and TLR4(-/-) mice transplanted with TLR4(-/-) BM. Hepatic mRNA expression of fibrogenic markers (collagen α1(I), TIMP1, TGF-Ɵ1) and inflammatory cytokines (IL-1Ɵ, IL-6) were markedly increased in WT mice with WT BM, but there was less of an increase in both TLR4-chimeric mice and in TLR4(-/-) mice transplanted with TLR4(-/-) BM. CONCLUSIONS: TLR4 signaling in both BM-derived and non-BM-derived liver cells is required for liver steatosis, inflammation, and a fibrogenic response after chronic alcohol treatment.


Subject(s)
Bone Marrow/drug effects , Fatty Liver, Alcoholic/metabolism , Liver/metabolism , Toll-Like Receptor 4/metabolism , Animals , Bone Marrow/metabolism , Central Nervous System Depressants/blood , Central Nervous System Depressants/toxicity , Cytokines/biosynthesis , Ethanol/blood , Ethanol/toxicity , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Liver/cytology , Liver/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors , Triglycerides/analysis
7.
Hepatology ; 50(1): 185-97, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19441102

ABSTRACT

UNLABELLED: Chemokines and chemokine receptors contribute to the migration of hepatic stellate cells (HSCs) and Kupffer cells, two key cell types in fibrogenesis. Here, we investigate the role of CCR2, the receptor for monocyte chemoattractant protein (MCP)-1, MCP-2, and MCP-3, in hepatic fibrosis. Hepatic CCR2, MCP-1, MCP-2, and MCP-3 messenger RNA expression was increased after bile duct ligation (BDL). Both Kupffer cells and HSCs, but not hepatocytes, expressed CCR2. BDL- and CCl(4)-induced fibrosis was markedly reduced in CCR2(-/-) mice as assessed through collagen deposition, alpha-smooth muscle actin expression, and hepatic hydroxyproline content. We generated CCR2 chimeric mice by the combination of clodronate, irradiation, and bone marrow (BM) transplantation allowing full reconstitution of Kupffer cells, but not HSCs, with BM cells. Chimeric mice containing wild-type BM displayed increased macrophage recruitment, whereas chimeric mice containing CCR2(-/-) BM showed less macrophage recruitment at 5 days after BDL. Although CCR2 expressed in the BM enhanced macrophage recruitment in early phases of injury, CCR2 expression on resident liver cells including HSCs, but not on the BM, was required for fibrogenic responses in chronic fibrosis models. In vitro experiments demonstrated that HSCs deficient in CCR2(-/-) or its downstream mediator p47phox(-/-) did not display extracellular signal-regulated kinase and AKT phosphorylation, chemotaxis, or reactive oxygen species production in response to MCP-1, MCP-2, and MCP-3. CONCLUSION: Our results indicate that CCR2 promotes HSC chemotaxis and the development of hepatic fibrosis.


Subject(s)
Liver Cirrhosis/etiology , Receptors, CCR2/physiology , Animals , Mice , Mice, Inbred C57BL
8.
Article in English | MEDLINE | ID: mdl-12687724

ABSTRACT

Retinoic acid (RA) is produced via two sequential reactions of retinol dehydrogenases (RDHs) and retinal dehydrogenases (RALDHs). We found that primary cultured mouse hepatocytes on a single collagen gel gradually lost hepatocyte specific morphology, and that another collagen gel overlay remarkably recovered it. The levels of albumin and liver-dominant expressed RDHs expression in hepatocytes paralleled their morphological change, decreased during single collagen gel culture, and were up-regulated by sequential collagen overlay. Quite similar to the expression changes, albumin and those RDHs' mRNA expression levels increased along with liver differentiation during pre- and postnatal liver development. Our data supports that all-trans and 9-cis RA, catalyzed by the RDHs, indeed play an important role in liver differentiation and regeneration.


Subject(s)
Aldehyde Oxidoreductases/physiology , Hepatocytes/cytology , Hepatocytes/enzymology , Hydro-Lyases/physiology , Tretinoin/physiology , Aldehyde Oxidoreductases/metabolism , Animals , Catalysis , Cell Differentiation , Cells, Cultured , Collagen , Culture Media , Gels , Hepatocytes/metabolism , Hydro-Lyases/metabolism , Liver Regeneration , Male , Mice , Mice, Inbred BALB C , Retinal Dehydrogenase , Tretinoin/metabolism
9.
Fibrogenesis Tissue Repair ; 5(Suppl 1): S17, 2012.
Article in English | MEDLINE | ID: mdl-23259769

ABSTRACT

Most chronic liver diseases of all etiologies result in progressive liver fibrosis. Myofibroblasts produce the extracellular matrix, including type I collagen, which constitutes the fibrous scar in liver fibrosis. Normal liver has little type I collagen and no detectable myofibroblasts, but myofibroblasts appear early in experimental and clinical liver injury. The origin of the myofibroblast in liver fibrosis is still unresolved. The possibilities include activation of endogenous mesenchymal cells including fibroblasts and hepatic stellate cells, recruitment from the bone marrow, and transformation of epithelial or endothelial cells to myofibroblasts. In fact, the origin of myofibroblasts may be different for different types of chronic liver diseases, such as cholestatic liver disease or hepatotoxic liver disease. This review will examine our current understanding of the liver myofibroblast.

10.
Hepatol Res ; 41(7): 683-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21711427

ABSTRACT

Non-alcoholic steatohepatitis (NASH) represents the progression of hepatic steatosis to streatohepatitis, fibrosis and cirrhosis. Three signaling pathways have been associated with this progression; Toll-like receptors, reactive oxygen species and Jun N-terminal kinase. This review will describe how activation of these three pathways is required for development of fibrosis in murine models of NASH. The three pathways are related and synergistic through intracellular cross-talk. Disruption of any of these pathways may inhibit NASH-induced fibrosis and are potential targets for therapeutic intervention.

11.
J Clin Invest ; 119(7): 1858-70, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19603542

ABSTRACT

Hepatic fibrosis develops as a response to chronic liver injury and almost exclusively occurs in a proinflammatory environment. However, the role of inflammatory mediators in fibrogenic responses of the liver is only poorly understood. We therefore investigated the role of CC chemokines and their receptors in hepatic fibrogenesis. The CC chemokines MIP-1alpha, MIP-1beta, and RANTES and their receptors CCR1 and CCR5 were strongly upregulated in 2 experimental mouse models of fibrogenesis. Neutralization of CC chemokines by the broad-spectrum CC chemokine inhibitor 35k efficiently reduced hepatic fibrosis, and CCR1- and CCR5-deficient mice displayed substantially reduced hepatic fibrosis and macrophage infiltration. Analysis of fibrogenesis in CCR1- and CCR5-chimeric mice revealed that CCR1 mediates its profibrogenic effects in BM-derived cells, whereas CCR5 mediates its profibrogenic effects in resident liver cells. CCR5 promoted hepatic stellate cell (HSC) migration through a redox-sensitive, PI3K-dependent pathway. Both CCR5-deficient HSCs and CCR1- and CCR5-deficient Kupffer cells displayed strong suppression of CC chemokine-induced migration. Finally, we detected marked upregulation of RANTES, CCR1, and CCR5 in patients with hepatic cirrhosis, confirming activation of the CC chemokine system in human fibrogenesis. Our data therefore support a role for the CC chemokine system in hepatic fibrogenesis and suggest distinct roles for CCR1 and CCR5 in Kupffer cells and HSCs.


Subject(s)
Liver Cirrhosis, Experimental/etiology , Receptors, CCR1/physiology , Receptors, CCR5/physiology , Animals , CCR5 Receptor Antagonists , Cell Movement , Humans , Kupffer Cells/physiology , Liver Cirrhosis, Experimental/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, CCR1/antagonists & inhibitors , Receptors, CCR1/genetics , Receptors, CCR5/genetics
12.
Microbiol Immunol ; 52(11): 513-21, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19090830

ABSTRACT

Two methods of TSS diagnosis were evaluated: comparison of symptoms with clinical criteria and monitoring for evidence of selective activation of Vbeta2(+) T cells by the causative toxin, TSS toxin-1 (TSST-1). Ten patients with acute and systemic febrile infections caused by Staphylococcus aureus were monitored for increase in TSST-1-reactive Vbeta2(+) T cells during their clinical courses. Nine of the ten patients were diagnosed with TSS based on evidence of selective activation of Vbeta2(+) T cells by TSST-1; however, clinical symptoms met the clinical criteria for TSS in only six of these nine patients. In the remaining patient, clinical symptoms met the clinical criteria, but selective activation of Vbeta2(+) T cells was not observed. Time taken to reach the diagnosis of TSS could be significantly shortened by utilizing the findings from tracing Vbeta2(+) T cells. In vitro studies showed that TSST-1- reactive T cells from TSS patients were anergic in the early phase of their illness. Examining selective activation of Vbeta2(+) T cells could be a useful tool to supplement clinical criteria for early diagnosis of TSS.


Subject(s)
Shock, Septic/diagnosis , Staphylococcal Infections/diagnosis , T-Lymphocyte Subsets/metabolism , Adult , Bacterial Toxins/immunology , Enterotoxins/immunology , Female , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Male , Methicillin-Resistant Staphylococcus aureus/immunology , Middle Aged , Shock, Septic/immunology , Shock, Septic/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Superantigens/immunology , T-Lymphocyte Subsets/immunology
13.
Am J Physiol Gastrointest Liver Physiol ; 287(3): G695-705, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15044178

ABSTRACT

Acute ethanol exposure induces oxidative stress and apoptosis in primary rat hepatocytes. Previous data indicate that the mitochondrial permeability transition (MPT) is essential for ethanol-induced apoptosis. However, the mechanism by which ethanol induces the MPT remains unclear. In this study, we investigated the role of Bax, a proapoptotic Bcl-2 family protein, in acute ethanol-induced hepatocyte apoptosis. We found that Bax translocates from the cytosol to mitochondria before mitochondrial cytochrome c release. Bax translocation was oxidative stress dependent. Mitochondrial Bax formed a protein complex with the mitochondrial voltage-dependent anion channel (VDAC). Prevention of Bax-VDAC interactions by a microinjection of anti-VDAC antibody effectively prevented hepatocyte apoptosis by ethanol. In conclusion, these data suggest that Bax translocation from the cytosol to mitochondria leads to the subsequent formation of a Bax-VDAC complex that plays a crucial role in acute ethanol-induced hepatocyte apoptosis.


Subject(s)
Apoptosis/drug effects , Ethanol/pharmacology , Hepatocytes/drug effects , Hepatocytes/physiology , Ion Channel Gating/physiology , Ion Channels/physiology , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/physiology , Animals , Caspases/metabolism , Cell Membrane Permeability/drug effects , Cross-Linking Reagents , Cyclosporine/pharmacology , Cytochromes c/metabolism , Electrophysiology , Immunoblotting , Immunohistochemistry , Immunosuppressive Agents/pharmacology , In Vitro Techniques , Ion Channel Gating/drug effects , Ion Channels/drug effects , Male , Microinjections , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Oxidative Stress/physiology , Precipitin Tests , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein
14.
Gastroenterology ; 126(3): 873-85, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14988841

ABSTRACT

BACKGROUND & AIMS: Treatment of steatosis is important in preventing development of fibrosis in alcoholic liver diseases. This study aimed to examine if pioglitazone, an antidiabetic reagent serving as a ligand of peroxisome proliferator-activated receptor gamma (PPAR gamma), could prevent alcoholic fatty liver. METHODS: Rats fed with an ethanol-containing liquid diet were given the reagent at 10 mg/kg per day intragastrically for 6 weeks. Hepatic genes involved in actions of the reagent were mined by transcriptome analyses, and their changes were confirmed by real-time polymerase chain reaction and Western blotting analyses. The direct effects of pioglitazone on primary-cultured hepatocytes were also assessed in vitro. RESULTS: Pioglitazone significantly attenuated steatosis and lipid peroxidation elicited by chronic ethanol exposure without altering insulin resistance. Mechanisms for improving effects of the reagent appeared to involve restoration of the ethanol-induced down-regulation of c-Met and up-regulation of stearoyl-CoA desaturase (SCD). Such effects of pioglitazone on the c-Met signaling pathway resulted from its tyrosine phosphorylation and resultant up-regulation of the apolipoprotein B (apoB)-mediated lipid mobilization from hepatocytes through very low-density lipoprotein (VLDL) as well as down-regulation of sterol regulatory element binding protein (SREBP) -1c and SCD levels and a decrease in triglyceride synthesis in the liver. CONCLUSIONS: Pioglitazone activates c-Met and VLDL-dependent lipid retrieval and suppresses triglyceride synthesis and thereby serves as a potentially useful stratagem to attenuate ethanol-induced hepatic steatosis.


Subject(s)
Fatty Liver, Alcoholic/prevention & control , Proto-Oncogene Proteins c-met/metabolism , Thiazolidinediones/pharmacology , Transcription Factors , Alcohol Drinking , Animals , Apolipoproteins B/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , DNA/biosynthesis , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Hepatocytes/drug effects , Hepatocytes/metabolism , Isoenzymes/metabolism , Lipid Peroxidation/drug effects , Liver/metabolism , Male , Oligonucleotide Array Sequence Analysis , Phosphorylation/drug effects , Pioglitazone , Proto-Oncogene Proteins c-met/genetics , Rats , Rats, Sprague-Dawley , Stearoyl-CoA Desaturase/metabolism , Sterol Regulatory Element Binding Protein 1 , Time Factors , Tyrosine/metabolism , Up-Regulation
15.
Am J Physiol Gastrointest Liver Physiol ; 287(5): G1078-85, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15475485

ABSTRACT

Oxidative stress is stated to be a central mechanism of hepatocellular injury in alcohol-induced liver injury. Recent reports have shown that Kupffer cell dysfunction in the leptin-deficient state contributes partly to the increased sensitivity to endotoxin liver injury. Here we report that leptin also plays a key role in the development of alcoholic liver injury and that leptin signaling in hepatocytes is involved in cellular mechanisms that mediate ethanol-induced oxidative stress. We found that chronic ethanol feeding in leptin receptor-deficient Zucker (fa/fa) rats for 6 wk resulted in a much more severe liver injury and augmented accumulation of hepatic lipid peroxidation compared with control littermates. The hepatic induction of stress-response and antioxidant proteins, such as metallothionein (MT)-1 and -2, was significantly suppressed in fa/fa rats after chronic ethanol feeding. Zinc concentration in liver was also decreased in fa/fa rats, compared with control littermates. In primary cultured hepatocytes from fa/fa rats, incubation with ethanol significantly suppressed MT-1 and -2 expressions. Addition of leptin to leptin-deficient ob/ob mouse primary hepatocytes led to an increase in MT-1 and -2 mRNA levels and a decrease in oxidative stress after incubation with ethanol. In conclusion, leptin deficiency enhances sensitivity of rats to alcohol-induced steatohepatitis through hepatocyte-specific interaction of MT-1 and -2 and resultant exaggeration of oxidative stress in hepatocytes. These findings suggest that leptin resistance in hepatocytes is an important mechanism of alcohol-induced liver injury.


Subject(s)
Ethanol , Fatty Liver/chemically induced , Leptin/deficiency , Metallothionein/antagonists & inhibitors , Animals , Drug Administration Schedule , Ethanol/administration & dosage , Fatty Liver/pathology , Fluorescent Dyes , Immunohistochemistry , Kupffer Cells/drug effects , Kupffer Cells/metabolism , Male , Metallothionein/genetics , Metallothionein/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/metabolism , Rats , Rats, Zucker , Tumor Necrosis Factor-alpha/genetics , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL