Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
J Hepatol ; 55(6): 1281-90, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21703211

ABSTRACT

BACKGROUND & AIMS: Cytosolic phospholipase A(2)α (cPLA(2)α) is a rate-limiting key enzyme controlling the release of arachidonic acid (AA) substrate for the synthesis of prostaglandins and leukotrienes. This study was designed to explore the role of hepatocyte cPLA(2)α in Fas-mediated liver injury, in vivo. METHODS: Transgenic mice with targeted expression of cPLA(2)α under control of the albumin-promoter enhancer and wild-type mice were injected intraperitoneally with anti-Fas antibody Jo2 or lipopolysaccharide plus d-galactosamine and monitored for liver injury and survival at various time points. RESULTS: The cPLA(2)α Tg mice resist Fas-induced liver failure, as reflected by the lower serum transaminase levels, fewer apoptotic hepatocytes, reduced caspase activation, and reduced PARP cleavage when compared to the matched wild type mice. Inhibition of cPLA(2)α by its pharmacological inhibitor, pyrrolidine, enhanced Jo2-induced liver injury in both cPLA(2)α Tg and wild type mice. Hepatic overexpression of cPLA(2)α increases the expression of EGFR in the liver and the EGFR inhibitor, AG1478, exacerbated Jo2-mediated liver injury. The cPLA(2)α transgenic mice develop more prominent liver tissue damage than wild-type mice after LPS/d-galactosamine injection. CONCLUSIONS: Hepatocyte cPLA(2)α protects against Fas-induced liver injury and this effect is mediated at least in part through the upregulation of EGFR.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Group IV Phospholipases A2/metabolism , Animals , Apoptosis/drug effects , Caspases/metabolism , Chemical and Drug Induced Liver Injury/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Galactosamine/toxicity , Gene Expression , Group IV Phospholipases A2/antagonists & inhibitors , Group IV Phospholipases A2/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Quinazolines/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tyrphostins/pharmacology , fas Receptor/toxicity
2.
Toxicol Appl Pharmacol ; 231(3): 336-43, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18572215

ABSTRACT

Fas stimulation recruits neutrophils and activates macrophages that secrete tumor necrosis factor-alpha (TNF-alpha), which aggravates Fas-mediated liver injury. To determine whether nonsteroidal anti-inflammatory drugs modify these processes, we challenged 24-hour-fasted mice with the agonistic Jo2 anti-Fas antibody (4 microg/mouse), and treated the animals 1 h later with saline or ibuprofen (250 mg/kg), a dual cyclooxygenase (COX)-1 and COX-2 inhibitor. Ibuprofen attenuated the Jo2-mediated recruitment/activation of myeloperoxidase-secreting neutrophils/macrophages in the liver, and attenuated the surge in serum TNF-alpha. Ibuprofen also minimized hepatic glutathione depletion, Bid truncation, caspase activation, outer mitochondrial membrane rupture, hepatocyte apoptosis and the increase in serum alanine aminotransferase (ALT) activity 5 h after Jo2 administration, to finally decrease mouse mortality at later times. The concomitant administration of pentoxifylline (decreasing TNF-alpha secretion) and infliximab (trapping TNF-alpha) likewise attenuated the Jo2-mediated increase in TNF-alpha, the decrease in hepatic glutathione, and the increase in serum ALT activity 5 h after Jo2 administration. The concomitant administration of the COX-1 inhibitor, SC-560 (10 mg/kg) and the COX-2 inhibitor, celecoxib (40 mg/kg) 1 h after Jo2 administration, also decreased liver injury 5 h after Jo2 administration. In contrast, SC-560 (10 mg/kg) or celecoxib (40 or 160 mg/kg) given alone had no significant protective effects. In conclusion, secondary TNF-alpha secretion plays an important role in Jo2-mediated glutathione depletion and liver injury. The combined inhibition of COX-1 and COX-2 by ibuprofen attenuates TNF-alpha secretion, glutathione depletion, mitochondrial alterations, hepatic apoptosis and mortality in Jo2-treated fasted mice.


Subject(s)
Apoptosis/physiology , Glutathione/deficiency , Hepatitis/metabolism , Ibuprofen/administration & dosage , Tumor Necrosis Factor-alpha/blood , fas Receptor/toxicity , Animals , Apoptosis/drug effects , Apoptosis/immunology , Hepatitis/enzymology , Hepatitis/mortality , Hepatitis/pathology , Hepatocytes/drug effects , Hepatocytes/pathology , Ibuprofen/pharmacology , Male , Mice , Mice, Inbred ICR , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
3.
Shock ; 30(2): 212-6, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18180697

ABSTRACT

Fulminant hepatic failure is a serious disease that has a poor cure rate unless liver transplantation is performed. Edaravone, a free radical scavenger, has been approved for the treatment of acute cerebral infarction, and its mechanism of action involves scavenging free radicals generated in ischemic tissues. We assessed the ability of 3-methyl-1-phenyl-2-pyrazolim-5-one (edaravone) to prevent Fas-induced acute liver failure in mice and examined the mechanisms underlying the observed effects. BALB/c mice were administered 0.25 microg/g (i.v.) body weight of a purified hamster agonist anti-Fas monoclonal antibody (clone Jo2). The mice also received either edaravone or isotonic sodium chloride solution before or after Jo2 treatment. Edaravone improved the survival rate of the mice markedly. Histopathological findings and serum aspartate aminotransferase levels showed that edaravone reduced the degree of liver injury caused by Jo2. Terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick end labeling staining showed that edaravone reduced the number of apoptotic hepatocytes. Edaravone also prevented cytochrome c release and caspase 3 activity, recognized as markers of apoptosis after mitochondrial disruption. Therefore, we considered that the antiapoptotic activity of edaravone involved blocking signals in the mitochondria-dependent pathway of Fas-induced apoptosis. Mitochondrial Bcl-xL and Bax, which form a channel in the mitochondrial membrane and, by their balance, regulate its permeability, are involved in mitochondrial disruption. Western blotting showed that the Bcl-xL-Bax ratio of the edaravone group was much higher than that of the control group. In conclusion, edaravone might protect hepatocytes from Fas-induced mitochondria-dependent apoptosis by regulating mitochondrial Bcl-xL and Bax.


Subject(s)
Antipyrine/analogs & derivatives , Liver Failure, Acute/prevention & control , Mitochondria, Liver/drug effects , Mitochondrial Proteins/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , fas Receptor/toxicity , Animals , Antipyrine/therapeutic use , Edaravone , Female , Free Radical Scavengers/therapeutic use , Liver Failure, Acute/etiology , Liver Failure, Acute/mortality , Mice , Mice, Inbred BALB C , Mitochondria, Liver/metabolism , Mitochondrial Proteins/biosynthesis , Mitochondrial Proteins/physiology , Survival Analysis , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/physiology , bcl-X Protein/biosynthesis , bcl-X Protein/physiology , fas Receptor/antagonists & inhibitors
4.
Am J Respir Crit Care Med ; 176(6): 591-601, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17600273

ABSTRACT

RATIONALE: Indirect acute lung injury (ALI) is associated with high morbidity and mortality. No specific therapies have been developed, because the underlying pathophysiological processes remain elusive. OBJECTIVES: To investigate the contribution of Fas-induced apoptotic and nonapoptotic/inflammatory signaling to the pathology of indirect ALI. METHODS: A mouse model of indirect ALI, induced by successive exposure to hemorrhagic shock and cecal ligation and puncture, was used. Quantification of active caspase-3 and the short splice variant of FLICE-inhibitory protein, (FLIP)short, was performed by Western blotting and immunohistochemistry, and cytokines/chemokines were assessed by cytometric bead array or ELISA. M30 immunostaining was done to evaluate epithelial cell apoptosis. Lung injury was assessed on the basis of myeloperoxidase activity, bronchoalveolar lavage protein, and lung histology. MEASUREMENTS AND MAIN RESULTS: Twelve hours after insult, lung monocyte chemoattractant protein-1, keratinocyte-derived chemokine, macrophage inflammatory protein-2, IL-6, tumor necrosis factor-alpha, and caspase-3 were increased and FLIP(short) was decreased. Fas- and Fas ligand-deficient mice showed marked protection from lung inflammation and apoptosis and decreased ALI. This was associated with a 10-day survival benefit. Similarly, 4 hours after pulmonary instillation of Fas-activating antibody in vivo, lung chemokines were markedly elevated in background mice and, interestingly, to a similar degree in macrophage-deficient animals. Fas activation on lung epithelial cells in vitro led to chemokine production that was dependent on extracellular signal-regulated kinase. CONCLUSIONS: Activation of apoptotic and nonapoptotic/inflammatory Fas signaling is an early important pathophysiological event in the development of indirect ALI after hemorrhagic shock and sepsis, in which lung epithelial cells appear to play a central role.


Subject(s)
Apoptosis , Respiratory Distress Syndrome/pathology , fas Receptor/toxicity , Actins/metabolism , Animals , Antibodies, Monoclonal/toxicity , Blotting, Western , Bronchoalveolar Lavage Fluid/chemistry , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 3/metabolism , Cell Line , Cell Proliferation , Disease Models, Animal , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Mice, Mutant Strains , Prognosis , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/metabolism , Severity of Illness Index , fas Receptor/immunology
5.
Dig Dis Sci ; 52(9): 2396-402, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17415659

ABSTRACT

Hepatocyte apoptosis was recently described for NASH patients. The pathomechanisms are incompletely understood, but upregulation of the death receptor Fas was detectable on hepatocytes of NASH patients. We analyzed the sensitivity of fatty liver against CD95/Fas-mediated apoptotic cell death by injection of agonistic anti-Fas antibody (Jo2) in obese ob/ob mice and lean control animals. Ob/ob mice died within 12 hrs, whereas control animals survived. Liver enzymes were significantly increased compared to those in control mice (P < 0.001). Histological analysis and also TUNEL assay of liver sections from ob/ob mice exhibited massive liver injury. Activity of caspase 3 was significantly more enhanced in livers of ob/ob mice after Jo2 challenge. The increased sensitivity was confirmed in vitro by using ob/ob-derived primary hepatocytes. CD95 expression was similar in ob/ob and control mice. However, hepatocytes from ob/ob mice revealed a decreased mitochondrial membrane potential, suggesting that mitochondria play a potential role in this increased susceptibility.


Subject(s)
Apoptosis/immunology , Fatty Liver/pathology , Hepatocytes/pathology , fas Receptor/immunology , Animals , Apoptosis/drug effects , Caspases/metabolism , Cells, Cultured , Disease Models, Animal , Disease Progression , Fatty Liver/chemically induced , Fatty Liver/mortality , Hepatocytes/drug effects , Hepatocytes/metabolism , In Situ Nick-End Labeling , Leptin/deficiency , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/immunology , Mice , Mice, Inbred C57BL , Severity of Illness Index , Survival Rate , fas Receptor/biosynthesis , fas Receptor/toxicity
6.
Free Radic Biol Med ; 42(7): 971-84, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17349925

ABSTRACT

This study evaluated whether acute ethanol pretreatment potentiates Fas-mediated liver injury and if oxidative stress and CYP2E1 play a role in any enhanced hepatotoxicity. There were 3-fold increases of transaminases and more extensive apoptotic necrosis of hepatocytes and focal hemorrhages of the hepatic lobule in mice treated with Jo2 Fas agonistic antibody plus ethanol compared to saline control or to mice treated with Jo2 or ethanol alone. CYP2E1 catalytic activity and protein were increased 2-fold by the acute ethanol pretreatment. There were 2- and 2.5-fold increases of caspase-8 and caspase-3 activity and 1.6-fold increases of apoptotic-positive cells in the Jo2 plus acute ethanol group compared to the Jo2 alone group. Levels of TNF-alpha, malondialdehyde, 4-hydroxynonenal, protein carbonyl formation, 3-nitrotyrosine protein adducts, and inducible nitric oxide synthase were increased in the Jo2 plus ethanol group. The enhanced hepatotoxicity of Jo2 plus ethanol and the elevated oxidative stress and TNF levels were lower in CYP2E1 knockout mice compared to wild-type mice expressing CYP2E1 but higher than saline controls. Toxicity also declined in mice treated with gadolinium chloride, an inhibitor of the inducible nitric oxide synthase or the antioxidant, N-acetyl-L-cysteine. These data indicate that acute ethanol pretreatment is capable of elevating hepatic apoptosis and liver injury induced by Jo2 Fas agonistic antibody. The enhanced hepatotoxicity involves increased oxidative and nitrosative stress, and appears to be mediated by CYP2E1-dependent and also CYP2E1-independent mechanisms.


Subject(s)
Cytochrome P-450 CYP2E1/metabolism , Ethanol/administration & dosage , Liver/drug effects , Oxidative Stress , fas Receptor/toxicity , Animals , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , fas Receptor/metabolism
7.
J Clin Invest ; 116(9): 2493-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16955144

ABSTRACT

TNF-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family with potent apoptosis-inducing properties in tumor cells. In particular, TRAIL strongly synergizes with conventional chemotherapeutic drugs to induce tumor cell death. Thus, TRAIL has been proposed as a promising future cancer therapy. Little, however, is known regarding what the role of TRAIL is in normal untransformed cells and whether therapeutic administration of TRAIL, alone or in combination with other apoptotic triggers, may cause tissue damage. In this study, we investigated the role of TRAIL in Fas-induced (CD95/Apo-1-induced) hepatocyte apoptosis and liver damage. While TRAIL alone failed to induce apoptosis in isolated murine hepatocytes, it strongly amplified Fas-induced cell death. Importantly, endogenous TRAIL was found to critically regulate anti-Fas antibody-induced hepatocyte apoptosis, liver damage, and associated lethality in vivo. TRAIL enhanced anti-Fas-induced hepatocyte apoptosis through the activation of JNK and its downstream substrate, the proapoptotic Bcl-2 homolog Bim. Consistently, TRAIL- and Bim-deficient mice and wild-type mice treated with a JNK inhibitor were protected against anti-Fas-induced liver damage. We conclude that TRAIL and Bim are important response modifiers of hepatocyte apoptosis and identify liver damage and lethality as a possible risk of TRAIL-based tumor therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Liver/pathology , MAP Kinase Kinase 4/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Necrosis Factor-alpha/genetics , fas Receptor/toxicity , Animals , Apoptosis , Apoptosis Regulatory Proteins/deficiency , Bcl-2-Like Protein 11 , Cell Death , Crosses, Genetic , Enzyme Activation , Hepatocytes/cytology , Hepatocytes/physiology , Immunohistochemistry , Liver/drug effects , Membrane Glycoproteins/deficiency , Membrane Proteins/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Proto-Oncogene Proteins/deficiency , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/deficiency
8.
Hepatology ; 44(3): 592-601, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16941708

ABSTRACT

Fatty liver is vulnerable to conditions that challenge hepatocellular energy homeostasis. Lipid-laden hepatocytes highly express uncoupling protein-2 (UCP2), a mitochondrial carrier that competes with adenosine triphosphate (ATP) synthesis by mediating proton leak. However, evidence for a link between UCP2 expression and susceptibility of liver to acute injury is lacking. We asked whether absence of UCP2 protects ob/ob mice from Fas-mediated acute liver damage. UCP2-deficient ob/ob mice (ob/ob:ucp2-/-) and UCP2-competent littermates (ob/ob:ucp2+/+) received a single dose of agonistic anti-Fas antibody (Jo2). Low-dose Jo2 (0.15 mg/kg intraperitoneally) caused less serum alanine aminotransferase (ALT) elevation and lower apoptosis rates in ob/ob:ucp2-/- mice. High-dose Jo2 (0.40 mg/kg intraperitoneally) proved uniformly fatal; however, ob/ob:ucp2-/- mice survived longer with less depletion of liver ATP stores, indicating that fatty hepatocytes may benefit from lack of UCP2 during Jo2 challenge. Although UCP2 reportedly controls mitochondrial oxidant production, its absence had no apparent effect on fatty liver tissue malondialdehyde levels augmented by Jo2. This finding prompted us to determine UCP2 expression in Kupffer cells, a major source of intrahepatic oxidative stress. UCP2 expression was found diminished in Kupffer cells of untreated ob/ob:ucp2+/+ mice, conceivably contributing to increased oxidative stress in fatty liver and limiting the impact of UCP2 ablation. In conclusion, whereas UCP2 abundance in fatty hepatocytes exacerbates Fas-mediated injury by compromising ATP stores, downregulation of UCP2 in Kupffer cells may account for persistent oxidative stress in fatty liver. Our data support a cell-specific approach when considering the therapeutic effects of mitochondrial uncoupling in fatty liver disease.


Subject(s)
DNA/genetics , Fatty Liver/metabolism , Gene Expression , Membrane Transport Proteins/genetics , Mitochondrial Proteins/genetics , Acute Disease , Animals , Apoptosis , Disease Models, Animal , Fatty Liver/genetics , Fatty Liver/pathology , Female , Hepatocytes/metabolism , Hepatocytes/pathology , Ion Channels , Membrane Transport Proteins/metabolism , Mice , Mitochondrial Proteins/metabolism , Oxidative Stress , Polymerase Chain Reaction , Uncoupling Protein 2 , fas Receptor/toxicity
9.
Hepatology ; 44(1): 252-62, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16799968

ABSTRACT

The constitutive androstane receptor (CAR) modulates xeno- and endobiotic hepatotoxicity by regulating detoxification pathways. Whether activation of CAR may also protect against liver injury by directly blocking apoptosis is unknown. To address this question, CAR wild-type (CAR+/+) and CAR knockout (CAR-/-) mice were treated with the CAR agonist 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP) and then with the Fas agonist Jo2 or with concanavalin A (ConA). Following the administration of Jo2, hepatocyte apoptosis, liver injury, and animal fatalities were abated in TCPOBOP-treated CAR+/+ but not in CAR-/- mice. Likewise, acute and chronic ConA-mediated liver injury and fibrosis were also reduced in wild-type versus CAR(-/-) TCPOBOP-treated mice. The proapoptotic proteins Bak (Bcl-2 antagonistic killer) and Bax (Bcl-2-associated X protein) were depleted in livers from TCPOBOP-treated CAR+/+ mice. In contrast, mRNA expression of the antiapoptotic effector myeloid cell leukemia factor-1 (Mcl-1) was increased fourfold. Mcl-1 promoter activity was increased by transfection with CAR and administration of TCPOBOP in hepatoma cells, consistent with a direct CAR effect on Mcl-1 transcription. Indeed, site-directed mutagenesis of a putative CAR consensus binding sequence on the Mcl-1 promoter decreased Mcl-1 promoter activity. Mcl-1 transgenic animals demonstrated little to no acute liver injury after administration of Jo2, signifying Mcl-1 cytoprotection. In conclusion, these observations support a prominent role for CAR cytoprotection against Fas-mediated hepatocyte injury via a mechanism involving upregulation of Mcl-1 and, likely, downregulation of Bax and Bak.


Subject(s)
DNA/genetics , Hepatocytes/ultrastructure , Liver Diseases/prevention & control , Pyridines/pharmacology , Transcription Factors/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/drug effects , bcl-2-Associated X Protein/drug effects , Animals , Apoptosis/drug effects , Chemical and Drug Induced Liver Injury , Concanavalin A/toxicity , Constitutive Androstane Receptor , Gene Expression/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Immunohistochemistry , Liver Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron , Mutation , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/drug effects , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Polymerase Chain Reaction , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Cytoplasmic and Nuclear , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , fas Receptor/toxicity
10.
Hepatology ; 42(2): 400-10, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16025513

ABSTRACT

Cytochrome P450 2E1 (CYP2E1) may be a central pathway in generating oxidative stress, reactive oxygen species, and causing hepatotoxic injury by alcohol and various hepatotoxins. This study evaluated the ability of CYP2E1 to potentiate or synergize the hepatotoxicity of Fas in vivo. C57BL/6 mice were injected intraperitoneally with pyrazole (Pyr) to induce CYP2E1. Then, 16-hour fasted mice were administered agonistic Jo2 anti-Fas antibody ip. Other mice were treated with Pyr or Jo2 alone. Levels of serum aminotransferase were 8.3- and 6.3-fold higher in the Pyr/Jo2 group compared with Jo2 alone, respectively. Histological evaluation of liver showed more extensive acidophilic necrosis and severe pathological changes in the Pyr/Jo2-treated mice. DNA fragmentation and caspase-8 and -3 activities were more elevated in the Pyr/Jo2 group compared with Jo2 alone. CYP2E1 activity and protein levels were higher in the Pyr/Jo2 group than in Jo2 alone. Levels of inducible nitric oxide synthase, 3-nitrotyrosine protein adducts, malondialdehyde, and protein carbonyls were also higher in the Pyr/Jo2 group compared with Jo2 alone. Glutathione and activities of catalase and Cu-Zn superoxide dismutase were decreased in the Pyr/Jo2 group. Administration of chlormethiazole, an inhibitor of CYP2E1, to the Pyr/Jo2-treated mice caused a significant decrease of alanine aminotransferase and liver pathological changes in association with a decrease in CYP2E1 protein and activity. In conclusion, enhanced hepatotoxicity of Fas was found in mice with elevated levels of CYP2E1. We speculate that overexpression of CYP2E1 might synergize and increase the susceptibility to Fas induced-liver injury.


Subject(s)
Cytochrome P-450 CYP2E1/biosynthesis , Liver/pathology , fas Receptor/toxicity , Animals , Antioxidants/analysis , Apoptosis , Catalysis , Chlormethiazole/pharmacology , Enzyme Induction , Lipid Peroxidation , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type II , Pyrazoles/pharmacology
11.
J Immunol ; 172(2): 1304-10, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14707109

ABSTRACT

Recently, we have demonstrated that tumor-specific CD4+ Th cell responses can be rapidly induced in advanced melanoma patients by vaccination with peptide-loaded monocyte-derived dendritic cells. Most patients showed a T cell reactivity against a melanoma Ag 3 (MAGE-3) peptide (MAGE-3(243-258)), which has been previously found to be presented by HLA-DP4 molecules. To analyze the functional and specificity profile of this in vivo T cell response in detail, peptide-specific CD4+ T cell clones were established from postvaccination blood samples of two HLA-DP4 patients. These T cell clones recognized not only peptide-loaded stimulator cells but also dendritic cells loaded with a recombinant MAGE-3 protein, demonstrating that these T cells were directed against a naturally processed MAGE-3 epitope. The isolated CD4+ Th cells showed a typical Th1 cytokine profile upon stimulation. From the first patient several CD4+ T cell clones recognizing the antigenic peptide used for vaccination in the context of HLA-DP4 were obtained, whereas we have isolated from the second patient CD4+ T cell clones which were restricted by HLA-DQB1*0604. Analyzing a panel of truncated peptides revealed that the CD4+ T cell clones recognized different core epitopes within the original peptide used for vaccination. Importantly, a DP4-restricted T cell clone was stimulated by dendritic cells loaded with apoptotic or necrotic tumor cells and even directly recognized HLA class II- and MAGE-3-expressing tumor cells. Moreover, these T cells exhibited cytolytic activity involving Fas-Fas ligand interactions. These findings support that vaccination-induced CD4+ Th cells might play an important functional role in antitumor immunity.


Subject(s)
Antigens, Neoplasm/immunology , Dendritic Cells/transplantation , Epitopes, T-Lymphocyte/immunology , Immunotherapy, Adoptive , Melanoma/immunology , Melanoma/therapy , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Antigen Presentation , Antigens, Neoplasm/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Death/immunology , Cell Division/immunology , Cell Line , Cell Line, Tumor , Clone Cells , Cytokines/biosynthesis , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/metabolism , Fas Ligand Protein , HLA Antigens/metabolism , Humans , Immunotherapy, Adoptive/methods , Melanoma/pathology , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/toxicity , Molecular Sequence Data , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , fas Receptor/metabolism , fas Receptor/toxicity
12.
J Pharmacol Exp Ther ; 308(3): 1174-80, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14617692

ABSTRACT

Previously, we have shown that primary afferent neurons are necessary for disease activity in immune-mediated liver injury in mice. These nerve fibers are detectable by substance P (SP) immunocytochemistry in the portal tract of rodent liver. Antagonists of the neurokinin-1 receptor (NK-1R), which is the prime receptor of SP, prevented liver damage by suppressing the synthesis of proinflammatory cytokines. Here, we investigated the influence of primary afferent nerve fibers, SP, and NK-1 receptor antagonists on hepatocyte apoptosis in vivo induced by administration of activating anti-CD95 monoclonal antibody (mAb) to mice. Depletion of primary afferent nerve fibers by neonatal capsaicin treatment prevented CD95-mediated activation of caspase-3, measured as enzymatic activity in liver homogenates or by demonstration of hepatocellular immunoreactivity for active caspase-3 in liver slices, and liver damage. This effect was reversed by administration of SP to anti-CD95 mAb-treated mice depleted from primary afferent neurons. The presence of the NK-1R on mouse hepatocytes was demonstrated by immunocytochemistry and flow cytometry. Intraperitoneal pretreatment with the NK-1 receptor antagonists (2S,3S)-cis-2-(diphenylmethyl)-N-([2-methoxyphenyl]-methyl)-1-azabicyclo(2.2.2.)-octan-3-amine (CP-96,345) or (2S,3S)3-([3,5-bis(trifluoromethyl)phenyl]methoxy)-2-phenylpiperadine (L-733,060) dose dependently protected mice from CD95-mediated liver injury. Similar results were obtained when apoptotic liver damage was induced by administration of tumor necrosis factor-alpha to d-galactosamine-sensitized mice. In conclusion, SP, probably by binding to its receptor on hepatocytes, might aggravate apoptotic signals in these cells. Because NK-1 receptor antagonists not only suppress the proinflammatory cytokine response in the liver but also prevent liver cell apoptosis in vivo, they might be suitable drugs for treatment of immune-mediated liver disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Liver Diseases/prevention & control , Neurokinin-1 Receptor Antagonists , Protective Agents/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Apoptosis , Capsaicin/pharmacology , Chemical and Drug Induced Liver Injury , Dose-Response Relationship, Drug , Hepatocytes/metabolism , Mice , Mice, Inbred BALB C , Neurons, Afferent/physiology , Receptors, Neurokinin-1/metabolism , Tumor Necrosis Factor-alpha/toxicity , fas Receptor/immunology , fas Receptor/toxicity
13.
J Immunol ; 168(6): 2712-9, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11884437

ABSTRACT

B cell susceptibility to Fas-mediated apoptosis is regulated in a receptor-specific fashion. CD40 engagement produces marked sensitivity to Fas killing, whereas surface Ig (sIg) engagement blocks Fas signaling for cell death in otherwise sensitive, CD40-stimulated B cell targets, and thus, induces a state of Fas resistance. The signaling mediator, Bruton's tyrosine kinase (Btk), is required for certain sIg-triggered responses, and Btk is reported to directly bind Fas and block Fas-mediated apoptosis. For these reasons, the role of Btk as a mediator of sIg-induced Fas resistance was examined. Dysfunction of Btk through mutation, and absence of Btk through deletion did not interfere with induction of Fas resistance by anti-Ig. This may be due, at least in part, to induction of Btk-dependent Bcl-2 family members by anti-Ig after CD40 ligand treatment. However, the susceptibility to Fas-mediated apoptosis of B cell targets stimulated by CD40 ligand alone was increased in the absence of Btk. These results indicate that Fas resistance produced by sIg triggering does not require Btk, but suggests that in certain situations Btk modulates B cell susceptibility to Fas killing.


Subject(s)
B-Lymphocyte Subsets/enzymology , B-Lymphocyte Subsets/immunology , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/metabolism , fas Receptor/toxicity , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocyte Subsets/metabolism , Immunity, Innate/genetics , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, Antigen, B-Cell/physiology
14.
J Immunol ; 167(11): 6654-62, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11714837

ABSTRACT

Ag stimulation of CD8+ lymphocytes in vivo results in their migration to various tissues as well as the activation of a cytolytic program involving perforin, TNF-alpha, and Fas ligand. The liver is one of the main sites for infiltration by activated CD8+ T cells, and this is followed by the death of hepatocytes. The contribution of the various cytolytic components to this process is unclear. Hepatocyte damage by CD8+ T cells was studied using the MHC class I-restricted OVA-specific TCR transgenic mouse (OT-1) to examine the contribution of Fas to hepatocyte death. Activated CD8+ T cells from both OT-1 and Fas-deficient OT-1lpr mice migrated to the liver in similar numbers after OVA administration, but only in OT-1 mice was there evidence of significant hepatocyte damage histologically and by elevation of serum aspartate transaminase. These differences were not the result of inefficient induction of cytolytic activity in OT-1lpr liver T cells, since they were as cytolytic in vitro as OT-1 liver T cells. This was supported by findings of similar high levels of message for perforin, TNF-alpha, and Fas ligand in liver lymphocytes from both mice. These findings demonstrate that following Ag activation, infiltrating liver CD8+ T lymphocytes induce hepatocyte damage in a Fas-dependent manner.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Cytotoxicity, Immunologic/immunology , Liver/immunology , Liver/pathology , fas Receptor/physiology , Animals , Cell Death/immunology , Egg Proteins/administration & dosage , Egg Proteins/immunology , Female , Hepatocytes/immunology , Hepatocytes/pathology , Immunophenotyping , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/immunology , Species Specificity , Tumor Cells, Cultured , fas Receptor/toxicity
15.
Int Arch Allergy Immunol ; 121(3): 183-93, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10729776

ABSTRACT

BACKGROUND: Restimulation of T lymphocytes via the TCR/CD3 complex can result in CD95/CD95L-dependent activation-induced cell death (AICD). Although the correlation of AICD sensitivity to the T helper 1 phenotype was confirmed in different studies, the underlying mechanism is still debated. Thus, it has been suggested that in Th2 cells, AICD resistance is controlled by a TCR-induced upregulation of the CD95-associated inhibitory phosphatase, FAP-1. We and others demonstrated that AICD resistance is associated with a reduced surface expression of CD95L upon restimulation. METHODS: Utilizing RT-PCR, Western blotting and flow cytometry, we analyzed time-dependent changes in levels of CD95L mRNA, cytosolic protein and surface expression in five long-term human T cell clones and polarized helper populations. RESULTS: We confirm that the inducible CD95L surface expression is lower or absent in all tested AICD-resistant clones as compared to sensitive cells. It is of interest that striking differences with respect to the activation-dependent inducibility of CD95L mRNA expression in individual resistant clones were observed. In addition, alterations in the expression of the inhibitory phosphatase FAP-1 or TCR-dependent changes in CD95 sensitivity in AICD-resistant clones could be ruled out as a mechanism for AICD resistance of human T cell clones. CONCLUSIONS: (1) The data presented strongly support the previous notion that AICD resistance of human T cell clones is mainly regulated by a differential expression of CD95L. (2) Differential expression of CD95L on individual resistant clones results from a lack of mRNA induction in one set and from a markedly decreased surface expression of translated protein in another set of clones.


Subject(s)
Apoptosis/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , Antibodies, Monoclonal/toxicity , Carrier Proteins/immunology , Cell Polarity/immunology , Clone Cells/immunology , Coculture Techniques , Cytotoxicity, Immunologic , Fas Ligand Protein , Humans , Immunity, Innate , Jurkat Cells , Ligands , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Protein Tyrosine Phosphatases/immunology , RNA, Messenger/biosynthesis , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Receptor-CD3 Complex, Antigen, T-Cell/physiology , T-Lymphocyte Subsets/enzymology , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Tumor Cells, Cultured , fas Receptor/immunology , fas Receptor/toxicity
16.
J Immunol ; 163(3): 1274-81, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10415024

ABSTRACT

Activation-induced cell death is a process by which overactivated T cells are eliminated, thus preventing potential autoimmune attacks. Two known mediators of activation-induced cell death are Fas(CD95) ligand (FasL) and APO2 ligand (APO2L)/TNF-related apoptosis-inducing ligand (TRAIL). We show here that upon mitogenic stimulation, bioactive FasL and APO2L are released from the T cell leukemia Jurkat and from normal human T cell blasts as intact, nonproteolyzed proteins associated with a particulate, ultracentrifugable fraction. We have characterized this fraction as microvesicles of 100-200 nm in diameter. These microvesicles are released from Jurkat and T cell blasts shortly (

Subject(s)
Apoptosis/immunology , Lymphocyte Activation , Membrane Glycoproteins/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism , Amino Acid Sequence , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Cell-Free System/chemistry , Cell-Free System/immunology , Cell-Free System/metabolism , Cell-Free System/ultrastructure , Cytochalasin B/pharmacology , Endopeptidases , Fas Ligand Protein , Flow Cytometry , Humans , Hydrolysis , Jurkat Cells , Ligands , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/toxicity , Microscopy, Electron, Scanning Transmission , Molecular Sequence Data , Phytohemagglutinins/pharmacology , T-Lymphocytes/chemistry , T-Lymphocytes/ultrastructure , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/toxicity , Ultracentrifugation , Vacuoles/chemistry , Vacuoles/immunology , Vacuoles/metabolism , Vacuoles/ultrastructure , fas Receptor/toxicity
17.
Cell Immunol ; 179(2): 153-64, 1997 Aug 01.
Article in English | MEDLINE | ID: mdl-9268499

ABSTRACT

Naive CD4(+) T cells proliferate strongly in response to superantigens such as staphylococcal enterotoxin B (SEB). When these cells are rested and challenged a second time, they undergo activation-induced cell death (AICD). Fas/FasL interactions have been shown to mediate AICD, even though the level of Fas expression in the 2 degrees SEB responder populations is no higher than in the 1 degrees cultures. To determine whether the dissimilarity between the 1 degrees and 2 degrees cultures could be attributed to differences in FasL cytotoxic activity or in the sensitivity of the Fas apoptosis signaling pathway, we compared these parameters during the 1 degrees and 2 degrees responses of lpr and gld CD4+ T cells (which do not undergo AICD due to a lack of Fas and an inactive FasL, respectively) so that each parameter could be evaluated independently. The results demonstrate that 2 degrees responders both express a higher level of functional FasL and are more sensitive to FasL-mediated killing. These findings account for the differences between the 1 degrees and 2 degrees responses of CD4+ T cells to superantigen. In addition, we found that the apparent level of FasL-mediated cytotoxic activity in the 2 degrees lpr CD4+ T cell population is much higher than that of wild-type cells, suggesting that deficient Fas expression leads to inordinately high levels of FasL expression or subsaturation of FasL binding sites.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Lymphocyte Activation , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/toxicity , Superantigens/pharmacology , fas Receptor/biosynthesis , fas Receptor/toxicity , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Antigens, Surface/physiology , Apoptosis/immunology , CD4-Positive T-Lymphocytes/metabolism , Enterotoxins/immunology , Enterotoxins/pharmacology , Fas Ligand Protein , Kinetics , Ligands , Membrane Glycoproteins/genetics , Mice , Mice, Inbred MRL lpr , Protein Biosynthesis , RNA/biosynthesis , Staphylococcus aureus/immunology , Superantigens/immunology , Time Factors , fas Receptor/genetics
18.
J Immunol ; 158(5): 1999-2006, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9036942

ABSTRACT

The perforin-facilitated entry of granzymes in target cells is a major mechanism used by CTL to induce cell death. It has been reported that granzyme B can cleave and activate the apoptotic cysteine protease p32 (CPP32)/Yama and its homologues in vitro. However, the mechanism for granzyme-based cytolysis exerted by intact CTL remains unclear. In the present work, we have used anti-CD3 mAb-redirected lysis of Fas-negative L1210 cells by CTL clones as a model to study perforin/granzyme-based cytotoxicity separately from the contribution of the Fas/Fas ligand system. N-acetyl-Asp-Glu-Val-Asp aldehyde (Ac-DEVD-CHO), a specific inhibitor of CPP32-like proteases, completely prevented the former type of lysis in 3-h assays, but not in long-term (16-h) assays. A combination of Ac-DEVD-CHO and the granzyme A inhibitor IGA (7-(phenyl-ureido)-4-chloro-3-(2-isothioureidoethoxy)-isocoumarin) inhibited long-term cytolysis. 3,4-Dichloroisocoumarin, a serine-protease inhibitor that efficiently inhibits granzyme B and poorly inhibits granzyme A, had similar effects as Ac-DEVD-CHO on anti-CD3 mAb-redirected lysis of L1210 cells. On the other hand, Fas-based cytolysis exerted by the same CTL clones on Fas-transfected L1210 cells (L1210Fas) was inhibited completely by Ac-DEVD-CHO, irrespective of the incubation time. These results suggest that granzyme B- and Fas-based cytotoxicity exerted by CTL clones converge at the level of CPP32-like protease activation, while granzyme A acts via a different, still undefined, pathway. We also demonstrate that perforin/granzyme-based cytolysis occurs without increase in the cellular ceramide content, ruling out the contribution of the sphingomyelinase pathway to this mechanism of cell death.


Subject(s)
Caspases , Cysteine Endopeptidases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Cytotoxicity, Immunologic/drug effects , Serine Endopeptidases/toxicity , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/toxicity , Animals , Caspase 3 , Ceramides/metabolism , Clone Cells , Cytotoxicity Tests, Immunologic , Drug Combinations , Granzymes , Humans , Immunoglobulin A/pharmacology , Isoantigens/immunology , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Oligopeptides/pharmacology , Perforin , Pore Forming Cytotoxic Proteins
19.
J Immunol ; 158(5): 2303-9, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9036978

ABSTRACT

Fas ligand (FasL) is a type II membrane protein. Binding of FasL to its receptor, Fas, induces apoptosis. Matrix metalloproteinase cleaves the membrane-bound human FasL to yield the active soluble form. Here, we have produced a large amount of human soluble rFasL using the yeast, Pichia pastoris. The purified rFasL was found to be glycosylated and to exist as a trimer. The rFasL was effective in inducing apoptosis in a Fas-expressing T cell or a fibroblast cell line. The ID50 of rFasL for mouse Fas-expressing T cells was about 0.5 ng/ml. The killing process with rFasL was quick. That is, >80% Fas-expressing mouse cells were killed within 1 h by a saturation concentration of human rFasL. Intravenous administration of 500 microg of human rFasL had a lethal effect in mice. When the mice were pretreated with Propionibacterium acnes, the subsequent injection of 30 microg of human rFasL induced hepatic failure and killed the mice within 24 h. These results indicated that the soluble human FasL is active in inducing apoptosis in vitro and in vivo, and its deleterious effect may be strengthened in patients who are suffering from bacterial infection.


Subject(s)
Membrane Glycoproteins/toxicity , Propionibacterium acnes/immunology , Recombinant Proteins/toxicity , fas Receptor/toxicity , Animals , Cell Line , Cytotoxicity Tests, Immunologic , Fas Ligand Protein , Female , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/mortality , Humans , Injections, Intraperitoneal , Ligands , Liver Failure/etiology , Liver Failure/immunology , Liver Failure/mortality , Membrane Glycoproteins/administration & dosage , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Recombinant Proteins/biosynthesis , fas Receptor/administration & dosage , fas Receptor/genetics
20.
Eur J Immunol ; 27(2): 449-55, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9045916

ABSTRACT

Elimination of activated T and B cells by Fas-dependent apoptosis may contribute to the maintenance of peripheral tolerance. CD40 ligation was recently shown to up-regulate Fas expression and enhance susceptibility to Fas-mediated apoptosis in mouse splenic B cells. In the present study, we have investigated the regulation of Fas expression and Fas-triggered apoptotis in mouse peritoneal B-1 cells. B-1 cells expressed a similar level of CD40 as that on B-2 cells, and proliferated in response to a soluble CD40 ligand (CD40L)-CD8alpha chimeric protein, suggesting that CD40 on B-1 cells is functional. In contrast to B-2 cells, B-1 cells expressed Fas at only low levels in response to CD40L-CD8alpha alone or CD40L-CD8alpha + interleukin-4, and were resistant to Fas-mediated apoptosis following these treatments. While Fas expression could be induced in B-1 cells to a comparable level as that in B-2 cells by cross-linking CD40L-CD8alpha with an anti-CD8alpha antibody, the sensitivity to Fas-mediated apoptosis in B-1 cells was significantly reduced compared with B2 cells. These results suggest that peritoneal B-1 cells from normal mice have a lower susceptibility to Fas-mediated apoptosis and may distinguish B-1 from B-2 cells. Similarly, B-1 cells from the peritoneal cavity and spleen of autoimmune-prone NZB mice exhibited reduced susceptibility to Fas-mediated apoptosis relative to their B-2 counterparts. NZB splenic B-1 cells, however, were more susceptible to Fas-mediated apoptosis than NZB peritoneal B-1 cells. The results presented here raise the possibility that the reduced susceptibility to Fas-triggered apoptosis in B-1 cells might be an accelerating factor for the autoantibody production in NZB mice.


Subject(s)
Apoptosis/immunology , B-Lymphocyte Subsets/drug effects , fas Receptor/toxicity , Animals , Autoimmune Diseases/pathology , B-Lymphocyte Subsets/metabolism , CD40 Antigens/biosynthesis , CD40 Antigens/toxicity , CD40 Ligand , CD8 Antigens/toxicity , Cells, Cultured , Female , Ligands , Membrane Glycoproteins/toxicity , Mice , Mice, Inbred BALB C , Mice, Inbred NZB , Peritoneal Cavity/cytology , Spleen/cytology , fas Receptor/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL