Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Med Res Arch ; 10(9)2022 Oct.
Article in English | MEDLINE | ID: mdl-36865784

ABSTRACT

Tumor necrosis factor -alpha (TNFα) is strongly associated with fatty liver disease (i.e, hepatosteatosis). Cytokine production has been thought of as a consequence of hepatic lipid accumulation which becomes a critical factor in the development of chronic liver pathologies as well as insulin resistance. The purpose of this study was to test the hypothesis that TNFα directly regulates lipid metabolism in liver in the mutant peroxisome-proliferator activated receptor-alpha (PPARα-/-) mouse model with robust hepatic lipid accumulation. At 10 weeks of age, TNFα and TNF receptor 1 expression are increased in livers of PPARα-/- mice compared to wild type. PPARα-/- mice were then crossed with mice lacking the receptor for TNFα receptor 1 (TNFR1-/-). Wild type, PPARα-/-, TNFR1-/-, PPARα-/- x TNFR1-/- mice were housed on ad-libitum standard chow diet for up to 40 weeks. Increases in hepatic lipid and liver injury and metabolic disruption associated with PPARα ablation were largely blunted when PPARα-/- mice were crossed with TNFR1-/- mice. These data support the hypothesis that TNFR1 signaling is critical for accumulation of lipid in liver. Therapies that reduce pro-inflammatory responses, namely TNFα, could have important clinical implications to reduce hepatosteatosis and progression of severe liver disease.

2.
Biol Res ; 51(1): 5, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29448959

ABSTRACT

BACKGROUND: Peroxisome proliferator activated receptor alpha (PPARα), a regulator of enzymes involved in ß oxidation, has been reported to influence lymphocyte activation. The purpose of this study was to determine whether PPARα plays a role in T cell-mediated hepatitis induced by Concanavalin A (ConA). METHODS: Wild type (wt) or PPARα-deficient (PPARα-/-) mice were treated with ConA (15 mg/kg) by intravenous injection 0, 10 or 24 h prior to sacrifice and serum and tissue collection for analysis of tissue injury, cytokine response, T cell activation and characterization. RESULTS: Ten and 24 h following ConA administration, wt mice had significant liver injury as demonstrated by serum transaminase levels, inflammatory cell infiltrate, hepatocyte apoptosis, and expression of several cytokines including interleukin 4 (IL4) and interferon gamma (IFNγ). In contrast, PPARα-/- mice were protected from ConA-induced liver injury with significant reductions in serum enzyme release, greatly reduced inflammatory cell infiltrate, hepatocellular apoptosis, and IFNγ expression, despite having similar levels of hepatic T cell activation and IL4 expression. This resistance to liver injury was correlated with reduced numbers of hepatic natural killer T (NKT) cells and their in vivo responsiveness to alpha-galactosylceramide. Interestingly, adoptive transfer of either wt or PPARα-/- splenocytes reconstituted ConA liver injury and cytokine production in lymphocyte-deficient, severe combined immunodeficient mice implicating PPARα within the liver, possibly through support of IL15 expression and/or suppression of IL12 production and not the lymphocyte as the key regulator of T cell activity and ConA-induced liver injury. CONCLUSION: Taken together, these data suggest that PPARα within the liver plays an important role in ConA-mediated liver injury through regulation of NKT cell recruitment and/or survival.


Subject(s)
Cytokines/immunology , Galactosylceramides/immunology , Hepatitis, Autoimmune/etiology , Macrolides/toxicity , PPAR alpha/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hepatitis, Autoimmune/immunology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
3.
Biol. Res ; 51: 5, 2018. tab, graf
Article in English | LILACS | ID: biblio-888430

ABSTRACT

Abstract Background Peroxisome proliferator activated receptor alpha (PPARα), a regulator of enzymes involved in β oxidation, has been reported to influence lymphocyte activation. The purpose of this study was to determine whether PPARα plays a role in T cell-mediated hepatitis induced by Concanavalin A (ConA). Methods Wild type (wt) or PPARα-deficient (PPARα−/−) mice were treated with ConA (15 mg/kg) by intravenous injection 0, 10 or 24 h prior to sacrifice and serum and tissue collection for analysis of tissue injury, cytokine response, T cell activation and characterization. Results Ten and 24 h following ConA administration, wt mice had significant liver injury as demonstrated by serum transaminase levels, inflammatory cell infiltrate, hepatocyte apoptosis, and expression of several cytokines including interleukin 4 (IL4) and interferon gamma (IFNγ). In contrast, PPARα−/− mice were protected from ConA-induced liver injury with significant reductions in serum enzyme release, greatly reduced inflammatory cell infiltrate, hepatocellular apoptosis, and IFNγ expression, despite having similar levels of hepatic T cell activation and IL4 expression. This resistance to liver injury was correlated with reduced numbers of hepatic natural killer T (NKT) cells and their in vivo responsiveness to alpha-galactosylceramide. Interestingly, adoptive transfer of either wt or PPARα−/− splenocytes reconstituted ConA liver injury and cytokine production in lymphocyte-deficient, severe combined immunodeficient mice implicating PPARα within the liver, possibly through support of IL15 expression and/or suppression of IL12 production and not the lymphocyte as the key regulator of T cell activity and ConA-induced liver injury. Conclusion Taken together, these data suggest that PPARα within the liver plays an important role in ConA-mediated liver injury through regulation of NKT cell recruitment and/or survival.


Subject(s)
Animals , Male , Mice , T-Lymphocytes/immunology , Cytokines/immunology , Macrolides/toxicity , Hepatitis, Autoimmune/etiology , PPAR alpha/immunology , Galactosylceramides/immunology , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Hepatitis, Autoimmune/immunology , Disease Models, Animal , Real-Time Polymerase Chain Reaction , Mice, Inbred C57BL
4.
Am J Pathol ; 186(6): 1524-36, 2016 06.
Article in English | MEDLINE | ID: mdl-27102767

ABSTRACT

The role of Fas-mediated apoptosis and its effect on proinflammatory cytokine production in early alcoholic liver disease has not been addressed. Wild-type mice (C57Bl/6) or mice with a functional mutation in the Fas ligand (B6.gld) were given either high-fat control diet or ethanol diet by intragastric cannulation for 2 or 4 weeks. Liver injury, hepatic lipid accumulation, and proinflammatory cytokine production associated with chronic ethanol consumption were largely prevented in B6.gld mice compared with wild-type mice. Conversely, B6.gld mice given ethanol exhibited increases in collagen deposition, hepatic collagen gene expression, and profibrogenic cytokines (eg, transforming growth factor-ß and IL-13) and alterations in matrix remodeling proteins (eg, matrix metalloproteinases and tissue inhibitor of metalloproteinases) compared with wild-type mice. Hepatic F4/80(+) macrophage populations were increased significantly in B6.gld mice compared with wild-type mice; hepatic CD3(+) cell populations were not significantly different. Importantly, a shift toward the expression of M2/Th2 cytokines (eg, IL-4 and IL-13) after ethanol exposure was observed in B6.gld mice compared with classical M1 cytokine expression in wild-type mice under similar conditions. In isolated macrophages, stimulation of Fas receptor minimally enhances lipopolysaccharide-induced M1 cytokine production and significantly limits M2 cytokine production. These data support the hypothesis that Fas-mediated signaling is important for an early ethanol-induced proinflammatory response but limits the profibrogenic response, regulating collagen production in response to chronic ethanol.


Subject(s)
Fas Ligand Protein/metabolism , Liver Cirrhosis/pathology , Liver Diseases, Alcoholic/pathology , Macrophages/metabolism , fas Receptor/metabolism , Animals , Apoptosis/physiology , Blotting, Western , Disease Models, Animal , Immunohistochemistry , In Situ Nick-End Labeling , Liver Cirrhosis/metabolism , Liver Diseases, Alcoholic/metabolism , Macrophages/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Phenotype , Real-Time Polymerase Chain Reaction
5.
Mol Immunol ; 67(2 Pt B): 265-75, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26105806

ABSTRACT

The beginning stages of liver damage induced by various etiologies (i.e. high fat diet, alcohol consumption, toxin exposure) are characterized by abnormal accumulation of lipid in liver. Alterations in intracellular lipid transport, storage, and metabolism accompanied by cellular insult within the liver play an important role in the pathogenesis of liver disease, often involving a sustained inflammatory response. The intracellular lipid transporter, fatty acid binding protein 5 (FABP5), is highly expressed in macrophages and may play an important role in the hepatic inflammatory response after endotoxin exposure in mice. This study tested the hypothesis that FABP5 regulates macrophage response to LPS in male C57bl/6 (wild type) and FABP5 knockout mice, both in vitro and in vivo. Treatment with LPS revealed that loss of FABP5 enhances the number of hepatic F4/80(+) macrophages in the liver despite limited liver injury. Conversely, FABP5 knock out mice display higher mRNA levels of anti-inflammatory cytokines IL-10, arginase, YM-1, and Fizz-1 in liver compared to wild type mice. Bone marrow derived macrophages stimulated with inflammatory (LPS and IFN-γ) or anti-inflammatory (IL-4) mediators also showed significantly higher expression of anti-inflammatory/regulatory factors. These findings reveal a regulatory role of FABP5 in the acute inflammatory response to LPS-induced liver injury, which is consistent with the principle finding that FABP5 is a regulator of macrophage phenotype. Specifically, these findings demonstrate that loss of FABP5 promotes a more anti-inflammatory response.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Inflammation/metabolism , Macrophages/metabolism , Macrophages/pathology , Neoplasm Proteins/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Polarity/drug effects , Cell Separation , Fatty Acid-Binding Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Inflammation/pathology , Lipopolysaccharides/pharmacology , Liver/injuries , Liver/metabolism , Macrophages/drug effects , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Proteins/genetics , Neutrophil Infiltration/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Time Factors
6.
Transpl Int ; 27(7): 748-58, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24649805

ABSTRACT

Liver regeneration is vital for graft survival and adequate organ function. Smad activation regulates hepatocyte proliferation and macrophage function. The aim of the current study was to evaluate the impact of Smad3 signaling during liver regeneration in the mouse. Male C57Bl/6 wild-type (wt) mice or mice deficient in Smad3 (Smad3(-/-) ) were subjected to a 70% partial hepatectomy (pHx) or sham surgery and sacrificed 24, 42, or 48 h later. Tissue was analyzed for TGF-ß signaling, the mitogenic cytokine response [i.e., tumor necrosis factor alpha, TNF-α; interleukin (IL)-6], and liver regeneration. Partial hepatectomy stimulated a strong regenerative response measured by proliferating cell nuclear antigen-positive hepatocytes 42 and 48 h post-pHx in conjunction with an increased expression of IL-6, TNF-α, and Smad2/3 phosphorylation 24 h post-pHx in both hepatocytes and nonparenchymal cells. Surprisingly, Smad3 deficiency led to reduced hepatocyte proliferation 42 h post-pHx which recovered by 48 h, a process that correlated with and was preceded by significant reductions in IL-6 expression and signal transducer and activator of transcription 3 phosphorylation, and cyclin D1 induction 24 h post-pHx. Loss of Smad3 signaling suppresses the expression of key mitogenic cytokines and delays hepatocellular regeneration. Therapies directed at finely regulating Smad3 activation early within the regenerating liver may prove useful in promoting liver cell proliferation and restoration of liver mass.


Subject(s)
Interleukin-6/biosynthesis , Liver Regeneration/physiology , STAT3 Transcription Factor/metabolism , Smad3 Protein/physiology , Animals , Hepatectomy , Male , Mice, Inbred C57BL , Protein Inhibitors of Activated STAT/physiology , Signal Transduction/drug effects
7.
Amino Acids ; 44(3): 925-31, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23135224

ABSTRACT

It has been demonstrated that a wide variety of white blood cells and macrophages (i.e. Kupffer cells, alveolar and peritoneal macrophages and neutrophils) contain glycine-gated chloride channels. Binding of glycine on the receptor stimulates Cl(-) influx causing membrane hyperpolarization that prevents agonist-induced influx of calcium. Since platelet-aggregation is calcium-dependent, this study was designed to test the hypothesis that glycine would inhibit platelet aggregation. Rats were fed diets rich of glycine for 5 days, while controls received isonitrogenous valine. The bleeding time and ADP- and collagen-induced platelet aggregation were measured. Dietary glycine significantly increased bleeding time about twofold compared to valine-treated controls. Furthermore, the amplitude of platelet aggregation stimulated with ADP or collagen was significantly decreased in whole blood drawn from rats fed 2.5 or 5 % dietary glycine by over 50 %. Addition of glycine in vitro (1-10 mM) also blunted rat platelet aggregation in a dose-dependent manner. Strychnine, a glycine receptor antagonist, abrogated the inhibitory effect of glycine on platelet-aggregation in vitro suggesting the glycine works via a glycine receptor. Glycine also blunted aggregation of human platelets. Further, the glycine receptor was detected in both rat and human platelets by western blotting. Based on these data, it is concluded that glycine prevents aggregation of platelets in a dose-dependent manner via mechanisms involving a glycine receptor.


Subject(s)
Blood Platelets/physiology , Glycine/metabolism , Platelet Aggregation , Animals , Bleeding Time , Down-Regulation , Female , Humans , Rats , Rats, Sprague-Dawley , Receptors, Glycine/genetics , Receptors, Glycine/metabolism
8.
Mol Immunol ; 47(4): 756-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19942291

ABSTRACT

The production of superoxide from NADPH oxidase by macrophages in response to endotoxin (LPS) is an important innate immune response, yet it is not clear how LPS signals the activation of NADPH oxidase. The hypothesis is that LPS-induced src kinase and PI3 kinase (PI3K) facilitates the activation of p47(phox), the regulatory subunit of NADPH oxidase. In mouse macrophage RAW264.7 cells, inhibition of src tyrosine family kinases inhibited LPS-induced activation of NADPH oxidase, phosphorylation of p47(phox), activation of PI3K and phosphorylation of the TLR4. Moreover, inhibition of LPS-induced increases in intracellular calcium blunted src kinase activation, PI3K association with TLR4, as well as PI3 kinase activation. These data suggest that both src kinase and PI3 kinase are involved in LPS-induced NADPH oxidase activation. Importantly, these data suggest that LPS-induced src kinase activation is critical for PI3 kinase activation as well as TLR4 phosphorylation and is dependent upon LPS-induced increase in intracellular calcium. These signaling events fill critical gaps in our understanding of LPS-induced free radical production as well as may potentially responsible for the mechanism of innate immune tolerance or desensitization caused by steroids or ethanol.


Subject(s)
Lipopolysaccharides/pharmacology , NADPH Oxidases/metabolism , src-Family Kinases/metabolism , Animals , Calcium/metabolism , Cell Line , Chelating Agents/pharmacology , Enzyme Activation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Macrophages/drug effects , Macrophages/enzymology , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Superoxides/metabolism , Toll-Like Receptor 4/metabolism , src-Family Kinases/antagonists & inhibitors
9.
Hepatology ; 51(1): 130-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20034047

ABSTRACT

UNLABELLED: Hepatosteatosis is associated with increased expression of tumor necrosis factor alpha (TNF-alpha) and interleukin (IL)-12, major T helper (Th) 1 cytokines, and reduced hepatic natural killer T (NKT) cell numbers. The relationship between lipid accumulation, cytokine expression, and hepatic NKT cells is not known. This study was conducted to assess the role of IL-12 in the development of hepatic steatosis and its potential impact on liver NKT cells. Male C57Bl/6 wildtype (WT) and IL-12-deficient (IL-12(-/-)) mice were fed a choline-deficient diet (CDD) for 0, 10, or 20 weeks. CDD led to marked hepatosteatosis, reduced hepatic but not splenic NKT cell numbers and function, and increased hepatic expression of the T(h)1-type cytokines IL-12, interferon gamma (IFN-gamma), and TNF-alpha in WT mice. The absence of IL-12 resulted in similar CDD-induced hepatosteatosis, but preserved hepatic NKT cells and significantly reduced hepatic IFN-gamma and TNF-alpha expression. Treatment of CDD-fed mice with lipopolysaccharide led to a significant increase in hepatic IL-12 expression, and Kupffer cell (KC) depletion reduced liver IL-12 expression and restored NKT cells in CDD-induced fatty liver. Interestingly, KCs from CDD-fed mice failed to produce increased quantities of IL-12 upon activation in vitro when compared to similarly treated KCs from control fed mice, suggesting that secondary factors in vivo promote heightened IL-12 production. Finally, human livers with severe steatosis showed a substantial decrease in NKT cells. CONCLUSION: Hepatosteatosis reduces the numbers of hepatic NKT cells in a KC-and IL-12-dependent manner. Our results suggest a pivotal and multifunctional role of KC-derived IL-12 in the altered immune response in steatotic liver, a process that is likely active within human nonalcoholic fatty liver disease.


Subject(s)
Fatty Liver/immunology , Interleukin-12/physiology , Kupffer Cells/physiology , Natural Killer T-Cells/immunology , Animals , Choline Deficiency/immunology , Fatty Liver/pathology , Humans , Interferon-gamma/biosynthesis , Interleukin-12/biosynthesis , Kupffer Cells/immunology , Liver/pathology , Macrophages/metabolism , Male , Mice , Tumor Necrosis Factor-alpha/biosynthesis
10.
Hepatology ; 47(1): 113-26, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18023023

ABSTRACT

UNLABELLED: Transforming growth factor beta (TGFbeta) promotes hepatocellular apoptosis and suppresses hepatic lymphocyte responses in part through activation of Smad3. The purpose of the current study was to determine the importance of Smad3 signaling in an experimental model of autoimmune hepatitis induced by concanavalin A (ConA), a process involving T cell activation and hepatocellular apoptosis. C57Bl/6 wild-type (Wt) or Smad3-deficient (Smad3(-/-)) mice were injected intravenously with 15 mg/kg ConA or vehicle. Nine hours post ConA injection, Wt mice presented with severe hepatitis as assessed by increased liver transferases. This injury was associated with eosinophil accumulation and preceded at 3 hours post-injection by significant increases in hepatic T helper 1 (interferon gamma) and T helper 2 (interleukin-4) cytokine production. Absence of Smad3 significantly blunted hepatocellular injury 9 hours post ConA injection, which was associated with reduced early T helper 1 and T helper 2 cytokine production and eosinophil accumulation. Smad3(-/-) livers also showed significant reductions in hepatocellular apoptosis as assessed by terminal UTP nick-end labeling when compared to ConA-treated Wt mice in conjunction with reduced caspase 3 cleavage, which was likely mediated by a Smad3-dependent inhibition of the survival factor extracellular signal-regulated kinase 1/2. In vitro, Smad3(-/-) hepatocytes were resistant to TGFbeta-induced apoptosis, and this protection was dependent on extracellular signal-regulated kinase activation. CONCLUSION: Together, these results show, for the first time, the significance of Smad3 signaling in autoimmune hepatitis, underlining the control of Smad3-dependent TGFbeta signaling on proinflammatory cytokine production, eosinophil recruitment, and hepatocellular apoptosis. Interruption of this pathway could be beneficial clinically to limit acute fulminant liver pathologies.


Subject(s)
Apoptosis/physiology , Hepatitis, Autoimmune/metabolism , Smad3 Protein/metabolism , T-Lymphocytes/physiology , Transforming Growth Factor beta/metabolism , Animals , Caspase 3/metabolism , Cell Survival/physiology , Concanavalin A/administration & dosage , Cytokines/metabolism , Eosinophils/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Hepatocytes/physiology , Injections , Liver/immunology , Liver/metabolism , Liver/pathology , Lymphocyte Activation/physiology , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology
11.
World J Gastroenterol ; 13(25): 3478-86, 2007 Jul 07.
Article in English | MEDLINE | ID: mdl-17659695

ABSTRACT

AIM: To investigate the effects of heme oxygenase-1 (HO-1) against oxidant-induced injury caused by bile duct ligation (BDL). METHODS: Either cobalt protoporphyrin (CoPP), a HO-1 inducer, or saline were injected intraperitoneally in male SD-rats. Three days later, BDL or sham-operations were performed. Rats were sacrificed 3 wk after BDL and livers were harvested for histology. Fibrosis was evaluated by sirius red staining and image analysis. Alpha-smooth muscular actin, which indicates activation of stellate cells, was detected by immunohistochemical staining, and cytokine and collagen-Ialpha (Col-Ialpha) mRNA expression was detected using RNase protection assays. RESULTS: Serum alanine transaminase increased 8-fold above normal levels one day after BDL. Surprisingly, enzyme release was not reduced in rats receiving CoPP. Liver fibrosis was evaluated 3 wk after BDL and the sirius red-positive area was found to be increased to about 7.8%. However, in CoPP pretreated rats sirius red-positive areas were increased to about 11.7% after BDL. Collagen-Ialpha and TGF-beta mRNA increased significantly by BDL. Again, this effect was increased by HO-1 overexpression. CONCLUSION: Hepatic fibrosis due to BDL is not reduced by the HO-1 inducer CoPP. In contrast, HO-1 overexpression increases liver injury in rats under conditions of experimental chronic cholestasis.


Subject(s)
Cholestasis/complications , Cholestasis/enzymology , Heme Oxygenase-1/physiology , Liver Cirrhosis/etiology , Actins/analysis , Alanine Transaminase/blood , Animals , Bile Ducts , Chronic Disease , Immunohistochemistry , Ligation , Liver/enzymology , Male , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/genetics , Tumor Necrosis Factor-alpha/genetics
12.
Hepatology ; 46(1): 229-41, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17596893

ABSTRACT

UNLABELLED: The regeneration of liver tissue following transplantation is often complicated by inflammation and tissue damage induced by a number of factors, including ischemia and reperfusion injury and immune reactions to the donor tissue. The purpose of the current study is to characterize the effects of T cell-mediated hepatitis induced by concanavalin A (ConA) on the regenerative response in vivo. Liver regeneration following a partial (70%) hepatectomy (pHx) was associated with elevations in serum enzymes and the induction of key cell cycle proteins (cyclin D, cyclin E, and Stat3) and hepatocyte proliferation. The induction of T cell-mediated hepatitis 4 days before pHx increased serum enzymes 48 hours after pHx, reduced early cyclin D expression and Stat3 activation, and suppressed hepatocyte proliferation. This inhibition of proliferation was also associated with increased expression of p21, the activation of Smad2, the induction of transforming growth factor beta and interferon gamma expression, and reduced hepatic interleukin 6 production. Moreover, the ConA pretreatment increased the numbers of separate oval cell-like CD117(+) cells and hematopoietic-like Sca-1(+) cell populations 48 hours following pHx. The depletion of natural killer (NK) cells, an important component of the innate immune response, did not affect liver injury or ConA-induced impairment of hepatocyte proliferation but did increase the numbers of both CD117-positive and Sca-1-positive cell populations. Finally, splenocytes isolated from ConA-pretreated mice exerted cytotoxicity toward autologous bone marrow cells in an NK cell-dependent manner. CONCLUSION: T cell-mediated hepatitis alters early cytokine responses, reduces hepatocellular regeneration, and induces NK cell-sensitive oval cell and hematopoietic-like cell expansion following pHx.


Subject(s)
Hepatitis/pathology , Liver Regeneration/physiology , T-Lymphocytes/pathology , Animals , Cell Survival , Concanavalin A/toxicity , Genes, Reporter , Hepatitis/immunology , Hepatitis/physiopathology , Interferon-gamma/genetics , Interleukin-6/genetics , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Polymerase Chain Reaction , T-Lymphocytes/immunology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor-alpha/genetics
13.
Exp Biol Med (Maywood) ; 232(4): 571-80, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17392494

ABSTRACT

The extent to which extrahepatic cells participate in liver regeneration following transplantation is not known. Either full-size or reduced-size livers from wild-type mice were implanted into green fluorescent protein-positive (GFP(+)) transgenic recipient mice to determine whether regenerated liver contained host-derived GFP(+) hepatic cells. After reduced-size liver transplantation, GFP(+) cells were localized to the portal zone of the liver lobule. Interestingly, GFP(+) cells stained for CD117, a marker for progenitor cells, beginning 2 days after transplantation. A significant number of GFP(+) CD117(+) cells were identified in donor livers after 28 days. GFP(+) cells comprised nearly 9% of the donor liver 28 days after reduced-size liver transplant. Moreover, GFP(+) cells also expressed the hepatic progenitor cell marker A6 and novel marker hepatic-specific antigen (HSA), as well as stem cell antigen-1 (Sca-1). Interestingly, some GFP(-) cells also were stained for CD117 and A6, suggesting that both extrahepatic and intrahepatic stem cells were present and may have contributed to the regenerative response under these conditions. Reduced-size liver transplantation using GFP(+) transgenic mice supports the hypothesis that recipient-derived progenitor cells are present and may contribute to liver regeneration following transplantation.


Subject(s)
Antigens, Differentiation/metabolism , Liver Regeneration , Liver Transplantation , Stem Cells/metabolism , Animals , Male , Mice , Mice, Transgenic , Stem Cells/cytology , Transplantation Chimera/metabolism
14.
Transplantation ; 82(9): 1214-20, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17102774

ABSTRACT

BACKGROUND: Tumor necrosis factor (TNF)-alpha is a cytokine with pleiotropic effects on the liver. The predominant hepatic receptor for TNFalpha is TNF receptor-1 (TNFR1). TNFR1 mediates liver injury after ischemia/reperfusion but is also mitogenic during hepatic regeneration. This study investigated the role of graft and host TNFR1 in early graft injury after liver transplantation in mice. METHODS: Livers from TNFR1 deficient (TNFR1-/-) and wild type (WT) mice were transplanted into either TNFR1-/- or WT recipients in all four possible combinations after 12 hours of cold storage. After eight hours, alanine transferase (ALT), necrosis, TdT-mediated dUTP-digoxigenin nick-end labeling (TUNEL) staining, caspase-3 activation, and myeloperoxidase were determined. RESULTS: When TNFR1-/- livers were transplanted into either WT or TNFR1-/- recipients, ALT was twofold greater than when WT donor livers were used. Necrosis and TUNEL staining also increased twofold and sevenfold, respectively, after transplantation of TNFR1-/- donor livers compared to WT. By contrast, ALT and necrosis decreased when WT or TNFR1-/- livers were transplanted into TNFR1-/- hosts compared to WT, which was associated with decreased neutrophil infiltration. CONCLUSION: In conclusion, graft and recipient TNFR1 has opposing effects. Graft TNFR1 decreases graft injury, whereas recipient TNFR1 mediates an increase of injury associated with enhanced neutrophil infiltration. Cross-transplanting of knockout and wild-type livers provides a new means to investigate graft-host interactions during hepatic injury.


Subject(s)
Graft Rejection/immunology , Liver Transplantation , Receptors, Tumor Necrosis Factor, Type I/physiology , Alanine Transaminase/blood , Animals , Caspase 3/analysis , Caspase 3/metabolism , Graft Rejection/genetics , Graft Rejection/pathology , Liver/enzymology , Liver/immunology , Male , Mice , Mice, Mutant Strains , Neutrophil Infiltration , Receptors, Tumor Necrosis Factor, Type I/genetics
15.
Am J Physiol Gastrointest Liver Physiol ; 290(6): G1318-28, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16439470

ABSTRACT

Although it is clear that bile acid accumulation is the major initiator of fibrosis caused by cholestatic liver disease, endotoxemia is a common side effect. However, the depletion of hepatic macrophages with gadolinium chloride blunts hepatic fibrosis. Because endotoxin is a key activator of hepatic macrophages, this study was designed to test the hypothesis that LPS signaling through CD14 contributes to hepatic fibrosis caused by experimental cholestasis. Wild-type mice and CD14 knockout mice (CD14(-/-)) underwent sham operation or bile duct ligation and were killed 3 wk later. Measures of liver injury, such as focal necrosis, biliary cell proliferation, and inflammatory cell influx, were not significantly different among the strains 3 wk after bile duct ligation. Markers of liver fibrosis such as Sirius red staining, liver hydroxyproline, and alpha-smooth muscle actin expression were blunted in CD14(-/-) mice compared with wild-type mice after bile duct ligation. Despite no difference in lymphocyte infiltration, the macrophage/monocyte activation marker OX42 (CD11b) and the oxidative stress/lipid peroxidation marker 4-hydroxynonenal were significantly upregulated in wild-type mice after bile duct ligation but not in CD14(-/-) mice. Increased profibrogenic cytokine mRNA expression in the liver after bile duct ligation was significantly blunted in CD14(-/-) mice compared with the wild type. The hypothesis that LPS was involved in experimental cholestatic liver fibrosis was tested using mice deficient in LPS-binding protein (LBP(-/-)). LBP(-/-) mice had less liver injury and fibrosis (Siruis red staining and hydroxyproline content) compared with wild-type mice after bile duct ligation. In conclusion, these data demonstrate that endotoxin in a CD14-dependent manner exacerbates hepatic fibrogenesis and macrophage activation to produce oxidants and cytokines after bile duct ligation.


Subject(s)
Cholestasis/immunology , Cholestasis/pathology , Disease Models, Animal , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/immunology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Animals , Cholestasis/complications , Liver/immunology , Liver/pathology , Liver Cirrhosis/etiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology
16.
Hepatology ; 41(2): 336-44, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15660387

ABSTRACT

The occurrence of malignant tumors of the upper gastrointestinal tract and liver is, based largely on epidemiological evidence, causally related to the consumption of ethanol. It is widely recognized that oxidants play a key role in alcohol-induced liver injury; however, it is unclear how oxidants may be involved in DNA damage. We asked whether nicotinamide adenine dinucleotide phosphate oxidase, cytochrome P450 CYP2E1, or both are responsible for the production of DNA damage. The rodent Tsukamoto-French model of intragastric ethanol infusion was used. Wistar rats, Cyp2e1-, p47(phox)-null, and hCyp2e1 transgenic mice were used. The abundance of oxidative DNA adducts, mutagenic apurinic/apyrimidinic sites, and expression of base excision DNA repair genes was determined. In rats and wild-type mice, ethanol treatment for 4 weeks led to an increase in oxidative DNA damage and induction of expression of the base excision DNA repair genes that are known to remove oxidative DNA lesions. No increase in either of the endpoints was observed in ethanol-treated Cyp2e1-null mice, whereas the magnitude of response in p47(phox)-null mice and transgenic hCyp2e1 was identical to that in wild types. The increase in expression of DNA repair genes was completely abolished by treatment with the P450 inhibitor 1-aminobenzotriazole. In conclusion, the data support the hypothesis that oxidative stress to DNA is induced in liver by ethanol. Furthermore, although it was shown that nicotinamide adenine dinucleotide phosphate oxidase-derived oxidants are critical for the development of ethanol-induced liver injury, CYP2E1 is required for the induction of oxidative stress to DNA, and thus may play a key role in ethanol-associated hepatocarcinogenesis.


Subject(s)
Cytochrome P-450 CYP2E1/physiology , DNA Damage/physiology , Ethanol/pharmacology , Liver/drug effects , Liver/metabolism , NADPH Oxidases/physiology , Oxidative Stress/physiology , Animals , DNA Repair/genetics , Gene Expression , Mice , Mice, Transgenic , Rats , Rats, Wistar
17.
Am J Physiol Gastrointest Liver Physiol ; 287(2): G310-4, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15246965

ABSTRACT

Ethanol consumption is known to cause significant acute liver damage resulting in hepatic fibrosis and eventual cirrhosis when consumed chronically. The mechanism(s) by which ethanol exerts its damaging effects on the liver are not well understood; however, recent scientific investigation has begun to delineate the earliest events in alcoholic liver disease. From these studies, it is apparent that components of the innate immune system and, in particular, Kupffer cells, play a significant role in this process. It is also becoming clear that other parts of the immune system including T cells may also be responsible for mediating the devastating effects of chronic alcohol consumption on the liver. This review will highlight recent experiments demonstrating a role for the innate immune response in the initiation and progression of alcohol-induced liver hepatitis and subsequent organ damage.


Subject(s)
Alcoholic Intoxication/immunology , Ethanol/immunology , Hepatitis, Alcoholic/immunology , Animals , Antibody Formation , Disease Progression , Hepatitis, Alcoholic/therapy , Humans , Inflammation Mediators/metabolism , Killer Cells, Natural , Kupffer Cells/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Liver Cirrhosis/immunology , Liver Diseases, Alcoholic/immunology , Signal Transduction , T-Lymphocytes
18.
Hepatology ; 39(3): 721-31, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14999690

ABSTRACT

Tumor necrosis factor alpha (TNFalpha) has been shown to be both proapoptotic and mitogenic for hepatocytes and necessary for alcohol-induced liver injury. Ras, a known proto-oncogene, is very important in the regulation of cellular responses to TNFalpha. Therefore, the purpose of this study was to investigate the role of Ras in alcohol-induced pathogenesis. Male C57Bl/6 mice were fed ethanol or high-fat control diet via intragastric cannulation for 4 weeks. Ras activity was increased significantly after 4 weeks of ethanol and correlated with an increase in pathologic features. However, in mice deficient in the receptor-type 1 for TNFalpha (TNFR1(-/-)), ethanol-induced liver injury and the increase in Ras activity were significantly blunted compared with wild-type mice. Furthermore, it was demonstrated that H-, K-, and R-Ras isoforms were increased after ethanol exposure in wild-type mice. In TNFR1(-/-) mice, R-Ras activity remained elevated by ethanol, whereas H-Ras and K-Ras activity was blunted significantly under these conditions. Interestingly, hepatocellular proliferation, which was elevated approximately fivefold after 4 weeks of chronic ethanol in wild-type mice, was also blunted in TNFR1(-/-) mice given ethanol. Inhibition of Ras with adenovirus containing a dominant-negative Ras had no effect on ethanol-induced liver injury, but significantly blunted ethanol-induced hepatocyte proliferation by more than 50%. Overexpression of mitochondrial superoxide dismutase using recombinant adenovirus blunted lipid peroxidation and attenuated hepatic injury resulting from ethanol, but had no effect on Ras activation and hepatocyte proliferation caused by ethanol. In conclusion, these data support the hypotheses that hepatocellular oxidative stress leads to cell death and that TNFalpha-induced Ras activation is important in hepatic proliferation in response to ethanol-induced liver injury.


Subject(s)
Ethanol/administration & dosage , Hepatocytes/pathology , Tumor Necrosis Factor-alpha/metabolism , ras Proteins/metabolism , Adenoviridae/genetics , Aldehydes/metabolism , Animals , Cell Division/drug effects , Drug Administration Schedule , Genes, Dominant , Genetic Vectors , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Staining and Labeling , Superoxide Dismutase/genetics , Superoxide Dismutase/pharmacology , Tumor Necrosis Factor-alpha/deficiency , ras Proteins/antagonists & inhibitors , ras Proteins/genetics
19.
Free Radic Biol Med ; 35(12): 1568-81, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14680680

ABSTRACT

Cytochrome P450 (CYP) 2E1 is induced by ethanol and is postulated to be a source of reactive oxygen species during alcoholic liver disease. However, there was no difference in liver pathology and radical formation between wild-type and CYP2E1 knockout mice fed ethanol. Other CYP isoforms may contribute these effects if CYP2E1 is inhibited or absent. The purpose of this study was, therefore, to determine if blocking most of the P450 isoforms with 1-aminobenzotriazole (ABT; 100 mg/kg i.g.), has any effect on liver damage and oxidative stress due to alcohol in rats and mice. Male C57BL/6 mice and Wistar rats were fed either high-fat control or ethanol-containing enteral diet for 4 weeks. ABT had a significant inhibitory effect on many P450 isoforms independent of concomitant alcohol administration. However, ABT did not protect against liver damage due to alcohol in either species. Indices of oxidative stress and inflammation were also similar in livers from vehicle-treated and ABT-treated animals fed ethanol. In summary, suppression of P450 activity with ABT had no apparent effect on oxidative stress caused by alcohol in both rats and mice. These data support the hypothesis that oxidative stress and liver damage can occur independently of CYP activities in both rats and mice during early alcohol-induced liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury , Cytochrome P-450 CYP2E1 Inhibitors , Enzyme Inhibitors/pharmacology , Ethanol/toxicity , Oxidative Stress/drug effects , Triazoles/pharmacology , Alanine Transaminase/blood , Animals , Cytochrome P-450 CYP2E1/biosynthesis , Cytochrome P-450 CYP2E1/genetics , Electron Spin Resonance Spectroscopy , Enzyme Induction , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Rats , Rats, Wistar
20.
Mol Cancer Res ; 1(12): 871-81, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14573788

ABSTRACT

A number of reports have described the effects of oxidative stress on tumor growth. Therefore, these experiments were designed to test the hypothesis that overexpression of extracellular superoxide dismutase (ecSOD) would inhibit the growth of tumors arising from s.c. implantation of syngenic B16-F1 melanoma cells. C57BL/6 mice were infected i.m. with adenovirus containing either beta-galactosidase (Ad.lacZ) as control or the secreted extracellular isoform of SOD (Ad.ecSOD) 3 days before s.c. implantation of B16-F1 tumor cells. Serum SOD activity was elevated nearly approximately 5-fold over control animals. Two weeks after implantation, B16-F1 tumor size was 65% smaller in mice infected with Ad.ecSOD in comparison with mice infected with Ad.lacZ. However, the presence of SOD did not affect growth rates of B16-F1 cells in vitro. Consistent with smaller tumor volume, tumors from Ad.ecSOD-infected mice also expressed less vascular endothelial growth factor (VEGF). Moreover, in vitro studies using B16-F1 cells confirm that SOD blunts oxidant-dependent VEGF expression. Importantly, CD31 expression and vessel density were markedly reduced in tumors from Ad.ecSOD-infected mice compared with controls. These data suggest that tumor oxidative stress may facilitate tumor vascularization and thus promote tumor growth.


Subject(s)
Extracellular Space/enzymology , Melanoma, Experimental/enzymology , Muscle, Skeletal/enzymology , Superoxide Dismutase/metabolism , Adenoviridae , Animals , Blotting, Western , Catalase/metabolism , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Vectors , Glutathione Peroxidase/metabolism , Immunohistochemistry , In Vitro Techniques , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Oxidative Stress , Recombinant Proteins , Time Factors , Transduction, Genetic , Up-Regulation , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...