ABSTRACT
Liver transplantation is the only therapy of proven benefit in fulminant hepatic failure (FHF). Lipopolysaccharide (LPS), D-galactosamine (GalN)-induced FHF is a well established model of liver injury in mice. Toll-Like Receptor 4 (TLR4) has been identified as a receptor for LPS. The aim of this study was to investigate the role of TLR4 in FHF induced by D-GalN/LPS administration in mice. Wild type (WT) and TLR4 deficient (TLR4ko) mice were studied in vivo in a fulminant model induced by GalN/LPS. Hepatic TLR4 expression, serum liver enzymes, hepatic and serum TNF-α and interleukin-1Ć levels were determined. Apoptotic cells were identified by immunohistochemistry for caspase-3. Nuclear factor-kappaĆ (NF-κ Ć) and phosphorylated c-Jun hepatic expression were studied using Western blot analysis. All WT mice died within 24 hours after administration of GalN/LPS while all TLR4ko mice survived. Serum liver enzymes, interleukin-1Ć, TNF-α level, TLR4 mRNA expression, hepatic injury and hepatocyte apoptosis all significantly decreased in TLR4ko mice compared with WT mice. A significant decrease in hepatic c-Jun and IκB signaling pathway was noted in TLR4ko mice compared with WT mice. In conclusion, following induction of FHF, the inflammatory response and the liver injury in TLR4ko mice was significantly attenuated through decreased hepatic c-Jun and NF-κB expression and thus decreased TNF-α level. Down-regulation of TLR4 expression plays a pivotal role in GalN/LPS induced FHF. These findings might have important implications for the use of the anti TLR4 protein signaling as a potential target for therapeutic intervention in FHF.
Subject(s)
Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Liver/drug effects , Toll-Like Receptor 4/metabolism , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Disease Models, Animal , Down-Regulation , Galactosamine/pharmacology , Interleukin-1beta/analysis , Interleukin-1beta/blood , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Signal Transduction/drug effects , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/bloodABSTRACT
BACKGROUND: Bax expression is a prerequisite for retinal ganglion cell (RGC) apoptosis. Experimental studies have reported Bax protein upregulation following optic nerve transection. The stimuli that trigger apoptosis share a common executioner proteolysis cascade, including caspase-3 and poly-(adenosine diphosphate ribose) polymerase cleavage. This study sought to elucidate the role of the mitochondrial apoptotic pathway in RGCs using a Bax transgenic knockout mouse model. METHODS: The right optic nerves of 26 C57BL mice, 7 Bax, 7 Bax, and 12 Bax, were subjected to crush injury and analyzed for apoptosis and neuronal cell loss on days 1, 3, and 21. Levels of Bax, Bcl-2, and caspase-3 messenger RNA expression were determined with real-time polymerase chain reaction. RESULTS: Multiple apoptotic cells were detected in the retinas of the Bax and Bax mice at days 1 and 3, but not in the Bax mice. The Bax/Bcl-2 ratio was higher in the Bax than in the Bax mice on day 1 (1.33 and 0.83, respectively), with a trend toward an increase on day 3 (1.47 and 1.66, respectively); Bax/Bcl-X showed the same elevation on day 1 in the wild-type mice (1.34) but decreased on day 3 (0.8). Bax gene expression was undetectable in the Bax mice. Caspase-3 gene expression was higher in the Bax than in the Bax mice on day 1 and dropped toward baseline on day 3. The opposite trend was noted in the Bax mice. CONCLUSION: The lack of apoptosis combined with the reduction in proapoptotic genes in the Bax mice after injury compared to the Bax and Bax mice suggests that Bax plays a crucial role in the induction of apoptosis. Suppression of Bax expression may reduce retinal cell loss.
Subject(s)
Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/cytology , bcl-2-Associated X Protein/physiology , Animals , Apoptosis , Caspase 3/genetics , Cell Survival/physiology , Gene Expression , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Nerve Crush , Optic Nerve Injuries/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Retina/pathology , Retinal Ganglion Cells/metabolismABSTRACT
The only currently offered curative option for many patients with primary or secondary liver tumors is the resection of hepatic tumors. The aim of this study was to evaluate the role of recombinant human erythropoietin (rhEPO) in liver protection and regeneration after subtotal hepatectomy in rats. Rats undergoing 70% hepatectomy received an intraperitoneal injection of saline (control) or rhEPO (4 U/g) 30 minutes prior to resection. Liver function was assessed by the measurement of the international normalized ratio (INR) levels, and hepatic injury was assessed by serum alanine aminotransferase and aspartate aminotransferase levels. Hepatic apoptosis was assessed by intrahepatic caspase-3 activity and morphological criteria. The regeneration capacity of remnant livers was assessed over 7 days with the regenerated liver/body weight ratio, immunohistochemistry markers of cell proliferation (Ki-67) and angiogenesis (von Willebrand factor), and phosphorylated extracellular signal-regulated kinase signaling. Two and 4 days after subtotal hepatectomy, the regenerated liver/body weight ratio was significantly higher in animals treated with rhEPO versus the control group (P < 0.005). Serum liver enzymes and INR levels on days 2 and 4 post-hepatectomy were significantly lower in animals pretreated with rhEPO in comparison with the control group (P < 0.005). No statistically significant difference was noted in intrahepatic hepatic caspase-3 activity, immunohistochemistry for caspase-3, or a terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling assay between the hepatectomized groups. In the rhEPO-pretreated group, the mitotic index, Ki-67 and von Willebrand factor expression, and extracellular signal-regulated kinase activity were significantly higher on day 2 post-hepatectomy (P < 0.05) in comparison with the control group. In conclusion, rhEPO treatment may offer a unique beneficial dual-function strategy for hepatic protection and regeneration immediately after subtotal hepatectomy in rats.
Subject(s)
Erythropoietin/pharmacology , Hepatectomy/methods , Liver Regeneration/drug effects , Postoperative Complications/drug therapy , Reperfusion Injury/drug therapy , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Blotting, Western , Caspase 3/metabolism , Humans , Immunohistochemistry , In Situ Nick-End Labeling , International Normalized Ratio , Ki-67 Antigen/metabolism , Liver/drug effects , Liver/pathology , Liver/physiology , Male , Organ Size , Postoperative Complications/pathology , Rats , Rats, Wistar , Recombinant Proteins , Reperfusion Injury/pathology , von Willebrand Factor/metabolismABSTRACT
Ischemia-reperfusion injury (I/R) is the main cause of primary graft nonfunction. Our aim was to evaluate the effect of excessive versus acute administration of erythropoietin (EPO) in attenuating the hepatic injury induced by I/R in mice. The effect of segmental (70%) hepatic ischemia was evaluated in a transgenic mouse line with constitutive overexpression of human EPO cDNA and in wild-type (WT) mice. Mice were randomly allocated to 5 main experimental groups: (i) WT-sham, (ii) WT ischemia, (iii) WT ischemia + recombinant human erythropoietin (rhEPO), (iv) transgenic-sham, and (v) transgenic ischemia. The EPO-pretreated mice showed a significant reduction in liver enzyme levels and intrahepatic caspase-3 activity and fewer apoptotic hepatocytes (p < 0.05 for all) compared with the WT untreated I/R group. EPO decreased c-Jun N-terminal kinase (JNK) phosphorylation and nuclear factor-κB (NF-κB) expression during I/R. In transgenic I/R livers, baseline histology showed diffused hepatic injury, and no significant beneficial effect was noted between the WT untreated and the transgenic I/R mice. In conclusion, acute pretreatment with EPO in WT mice attenuated in vivo I/R liver injury. However, in excessive EPO overexpression, the initial liver injury abolished the beneficial effect of EPO. These findings have important implications for the potential use of acute EPO in I/R injury during liver transplantation.
Subject(s)
Erythropoietin/administration & dosage , Liver/blood supply , Reperfusion Injury/drug therapy , Animals , Apoptosis/drug effects , Caspase 3/metabolism , DNA, Complementary/genetics , Female , Hematocrit , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Phosphorylation , Random Allocation , Recombinant Proteins , Reperfusion Injury/metabolism , Reperfusion Injury/pathologyABSTRACT
We have previously found that uridine 5'-triphosphate (UTP) significantly reduced cardiomyocyte death induced by hypoxia via activating P2Y(2) receptors. To explore the effect of UTP following myocardial infarction (MI) in vivo we studied four groups: sham with or without LAD ligation, injected with UTP (0.44microg/kg i.v.) 30min before MI, and UTP injection (4.4microg/kg i.v.) 24h prior to MI. Left ventricular end diastolic area (LVEDA), end systolic area (LVESA) fractional shortening (FS), and changes in posterior wall (PW) thickness were performed by echocardiography before and 24h after MI. In addition, we measured different biochemical markers of damage and infarct size using Evans blue and TTC staining. The increase in LVEDA and LVESA of the treated animals was significantly smaller when compared to the MI rats (p<0.01). Concomitantly, FS was higher in groups pretreated with UTP 30min or 24h (56+/-14.3 and 36.7+/-8.2%, p<0.01, respectively). Ratio of infarct size to area at risk was smaller in the UTP pretreated hearts than MI rats (22.9+/-6.6, 23.1+/-9.1%, versus 45.4+/-7.6%, respectively, p<0.001). Troponin T and ATP measurements, demonstrated reduced myocardial damage. Using Rhod-2-AM loaded cardiomyocytes, we found that UTP reduced mitochondrial calcium levels following hypoxia. In conclusion, early or late UTP preconditioning is effective, demonstrating reduced infarct size and superior myocardial function. The resulting cardioprotection following UTP treatment post ischemia demonstrates a reduction in mitochondrial calcium overload, which can explain the beneficial effect of UTP.
Subject(s)
Myocardial Infarction/drug therapy , Uridine Triphosphate/pharmacology , Adenosine Triphosphate/analysis , Animals , Blood Pressure/drug effects , Calcium/metabolism , Cardiotonic Agents/pharmacology , Cells, Cultured , Creatine Kinase/blood , Echocardiography , Heart/drug effects , Heart/physiology , Heart/physiopathology , Heart Rate/drug effects , Hypoxia/metabolism , Male , Mitochondria/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Rats , Rats, Wistar , Troponin T/bloodABSTRACT
BACKGROUND: Liver transplantation is the only therapy of proven benefit in fulminant hepatic failure (FHF). Lipopolysaccharide (LPS), d-galactosamine (GalN)-induced FHF is a well-established model of liver injury in mice. Erythropoietin has a powerful tissue-protective effect in animal models. The aim of this study was to investigate the effect and mechanism of recombinant human erythropoietin (rhEPO) administration in FHF mice. METHODS: C57BL/6 (n=42) mice were studied in vivo in a fulminant model induced by GalN/LPS. rhEPO was administered 30 min after the induction of FHF. Serum liver enzymes and hepatic tumor necrosis factor (TNF)-α and interleukin (IL)-1Ć levels were determined. Histologic analysis was performed, and apoptotic cells were identified by immunohistochemistry for caspase-3. Nuclear factor (NF)-κB and c-Jun-N-terminal kinase (JNK) activation were studied using Western blot analysis. RESULTS: After the induction of FHF, all control mice died within 12 hr of GalN/LPS administration. However, 83% of mice that were administered rhEPO were alive 2 weeks later, and overall survival improved (Kaplan-Meier, P<0.001). The serum liver enzymes, hepatic TNF-α and IL-1Ć levels, liver histologic injury, and apoptotic hepatocytes were significantly reduced in FHF mice that were administered rhEPO compared with untreated mice. A significant decrease in hepatic NF-κB and JNK activation was noted in FHF rhEPO-treated mice compared with FHF untreated mice. CONCLUSIONS: The administration of rhEPO brought about increased survival and attenuation of the hepatic injury. This was associated with decreased hepatic NF-κB and JNK activation and thus TNF-α and IL-1Ć levels. These findings have important implications for the potential use of rhEPO in FHF.
Subject(s)
Erythropoietin/pharmacology , Liver Failure, Acute/drug therapy , Alanine Transaminase/blood , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/blood , Disease Models, Animal , Galactosamine/toxicity , Humans , Interleukin-1beta/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Kaplan-Meier Estimate , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Liver Failure, Acute/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Recombinant Proteins , Tumor Necrosis Factor-alpha/metabolismABSTRACT
Myocardial injury, developed after a period of ischemia/reperfusion (I/R) results in the destruction of functional heart tissue, this being replaced by scar tissue. Intracellular signaling pathways mediating cardiomyocyte death are partially understood and involve the activation of Ras. p38-MAPK, JNK and Mst-1 are downstream effectors of Ras protein. We hypothesized that S-farnesylthiosalicylic acid (FTS), a synthetic small molecule that detaches Ras from the inner cell membrane, consequently inhibiting Ras activity, reduces I/R myocardial injury in vitro and in vivo. Wistar rat hearts were isolated, mounted on the Langendorff apparatus and subjected to ischemia (30 min, 37 degrees C) and reperfusion. During the reperfusion period, the hearts were perfused with FTS (1 microM) solution or control buffer. Left anterior descending (LAD) ligation and subsequent reperfusion was performed in two groups of Wistar rats. Rats received 5mg/kg FTS or PBS according to two protocols: (A) FTS or PBS were administered daily 7 days prior, immediately before and 14 days (every other day) after LAD occlusion or (B) every other day for 14 days post-I/R. Hearts from FTS-treated rats (Langendorff) and FTS-treated rats (protocol A) showed a significant improvement in myocardial performance and smaller scar tissue compared with the PBS group. Infarct size in the FTS-treated group was 12.7+/-2% vs. 23.7+/-4% in the PBS-treated (in vitro) group and 17.3+/-2.5% vs. 36+/-7% compared with control I/R rats (in vivo) p<0.05. These effects may be associated with the down regulation of JNK as a short-term effector and with Mst-1 in the long-term remodeling process.
Subject(s)
Farnesol/analogs & derivatives , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Salicylates/pharmacology , ras Proteins/antagonists & inhibitors , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Farnesol/administration & dosage , Farnesol/pharmacology , Heart Function Tests , In Vitro Techniques , Male , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Inbred Lew , Rats, Wistar , Salicylates/administration & dosageABSTRACT
BACKGROUND AND AIM: Mitochondrial calcium overload triggers apoptosis and also regulates ATP production. ATP and uridine-5'-triphosphate (UTP) depletion from hepatic tissue after ischemia causes cell death. ATP and UTP binds to cell membranes of the hepatocytes through P2Y receptors. Our aim was to investigate the role of UTP on the hepatic injury induced by ischemia. METHODS: Isolated mouse livers were randomly divided into five groups: (1) control group; (2) ischemic group (90 min); (3) as group 2, but with the administration of UTP; (4) as group 2, but with the administration of suramin, a P2Y antagonist; and (5) as group 3, but with the simultaneous administration of suramin and UTP. RESULTS: There was a postischemic significant reduction in the release of liver enzymes in the animals pretreated with UTP, the intrahepatic caspase-3 activity was significantly decreased, and the intrahepatic ATP content increased compared with group 2 (ischemic untreated). UTP prevented intracellular Ca overload after hypoxia in hepatocyte cultures. In the UTP-treated groups, significantly fewer apoptotic hepatocyte cells were noted by weaker activation of caspase-3 and by the transferase-mediated dUTP nick end labeling assay. The administration of suramin prevented the beneficial effect of endogenous ATP. UTP treatment attenuated the degradation of IkappaBalpha (nuclear factor-kappaB inhibitor) by 80% during reperfusion with no effect on c-Jun N terminal kinase phosphorylation. CONCLUSION: The administration of UTP before induction of ischemia-reperfusion can attenuate hepatic injury. UTP administration decreased cytosolic Ca overload in hypoxic conditions. UTP-mediated protective effects may be regulated through nuclear factor- kappaB inactivation. These findings have important implications for the potential use of UTP in ischemic hepatic injury.
Subject(s)
Liver/injuries , Reperfusion Injury/prevention & control , Uridine Triphosphate/therapeutic use , Alanine Transaminase/metabolism , Animals , Apoptosis/drug effects , Aspartate Aminotransferases/metabolism , Blood Pressure , Caspase 3/metabolism , Cell Hypoxia , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/physiology , L-Lactate Dehydrogenase/metabolism , Liver/cytology , Liver/drug effects , Liver/enzymology , Male , Mice , Suramin/therapeutic use , Vena Cava, Inferior/physiologyABSTRACT
INTRODUCTION: Apoptosis is a central mechanism of cell death following reperfusion of the ischemic liver. Recombinant human erythropoietin (rhEPO) have an important role in the treatment of myocardial ischemia/reperfusion (I/R) injury, by preventing apoptosis. The aim of the study was to investigate the effect of different regimens of rhEPO in preventing apoptosis following I/R-induced hepatic injury. MATERIAL AND METHODS: Isolated mouse livers were randomly divided into five groups: (1) control group, perfused for the whole study period (105 min); (2) 30-min perfusion followed by 90 min of ischemia and 15 min of reperfusion; (3), (4) and (5) like group 2, but with administration of rhEPO 5,000 units/kg i.p. at 30 min, 24 h, or both 30 min and 24 h respectively, before induction of ischemia. Perfusate liver enzyme levels and intrahepatic caspase-3 activity were measured, and apoptotic cells were identified by morphological criteria, TUNEL assay, and immunohistochemistry for caspase-3. Using immunoblot the expression of the proapoptotic JNK and inhibitor of NFkappaB (IkappaBalpha) were also evaluated. von Willebrand factor (vWF) immunohistochemistry was used as a marker of endothelial cells. RESULTS: Compared to the I/R livers, all 3 rhEPO pretreated groups showed: a significant reduction in liver enzyme levels (P < 0.05) and intrahepatic caspase-3 activity (P < 0.05), fewer apoptotic hepatocytes (P < 0.05) and positive vWF staining in numerous endothelial cells lining the sinusoids. EPO decreased JNK phosphorylation and the degradation of the inhibitor of NFkappaB (IkappaBalpha) during I/R. There was no added benefit of the multiple- over the single-dose rhEPO regimen. CONCLUSION: Pretreatment with one dose of rhEPO can attenuate post-I/R hepatocyte apoptotic liver damage. NFkappaB and JNK activation is likely to play a pivotal role in the pathophysiology of I/R hepatic injury and might have a key role in EPO-mediated protective effects. This effect is associated with the increase in sinusoidal vWF immunostaining suggests an additional effect of rhEPO in liver angiogenesis recovery. These findings have important implications for the potential use of rhEPO in I/R injury during liver transplantation.
Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Erythropoietin/pharmacology , Liver/blood supply , Liver/pathology , MAP Kinase Kinase 4/metabolism , Protein Serine-Threonine Kinases/metabolism , Reperfusion Injury/prevention & control , Animals , Disease Models, Animal , Humans , Ischemia , Liver/metabolism , Male , Mice , Random Allocation , Recombinant Proteins , Reperfusion , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , von Willebrand Factor/metabolism , NF-kappaB-Inducing KinaseABSTRACT
OBJECTIVES: The increased susceptibility to ischemic injury of hypertrophied hearts has long been recognized. The purpose of this study was to investigate the effects of pre-ischemic pharmacological preconditioning (PC) with adenosine A(1) or A(3) receptor activation, on the recovery of the isolated myocardium post cardioplegic ischemia. In addition, we examined the p38 MAPK activation in this process. MATERIALS AND METHODS: WKY and SHR hearts were subjected to two different modes of treatment. (1) In the perfusion mode- (the first window of PC) isolated rat hearts were perfused for 10 min with Krebs Henseleit solution and then A(1) receptor agonist (CCPA) or A(3) receptor agonist (Cl-IB-MECA), 10 nM for 20 min, followed by 30 min of warm cardioplegic ischemia and 30 min of reperfusion. (2) In the injection mode (the second window of PC) 100 microg/kg CCPA or Cl-IB-MECA, were administered 24 h before the experiment. Isolated hearts were perfused for 30 min with KH and then subjected to the same protocol as described above. RESULTS: Recovery of hemodynamic parameters was always better in the normal vs. hypertrophied hearts. CCPA improved recovery of left ventricular developed pressure, coronary flow and ATP levels of the hearts (normal and hypertrophied) in both modes of treatment. Cl-IB-MECA was partially beneficial especially in the injected mode. Increased phosphorylation of p38 MAPK relative to baseline, in both early (perfused) and late (injected) modes of treatment especially in the WKY hearts, is demonstrated. CONCLUSION: CCPA in both modes of treatment and Cl-IB-MECA, especially in the injected mode, were beneficial in protecting the normal and hypertrophied perfused isolated rat heart subjected to normothermic cardioplegic ischemia. This protection was partially related to the increased phosphorylation of p38 MAPK.
Subject(s)
Heart/drug effects , Ischemic Preconditioning, Myocardial/methods , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A3/metabolism , Reperfusion Injury/prevention & control , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A1 Receptor Agonists , Adenosine A3 Receptor Agonists , Analysis of Variance , Animals , Coronary Circulation/drug effects , Coronary Circulation/physiology , Heart/metabolism , Heart/physiology , Hypertrophy , Male , Rats , Rats, WistarABSTRACT
Apoptosis appears to be a central mechanism of cell death following reperfusion of the ischemic liver. The aim of this study was to determine the effect of decreased expression of the proapoptotic Bax gene on hepatic apoptotic warm ischemia/reperfusion (I/R) injury. Three groups of mice were studied: homozygotic knockout mice (Bax-/-); heterozygotic (Bax+/-); and wild type (Bax+/+). Isolated mouse livers were subjected to 90 minutes of ischemia (37 degrees C) followed by 15 minutes of reperfusion. Bax and Bcl-2 expression in liver tissue homogenates was measured by Western blot. Serum liver enzyme levels were measured and intrahepatic caspase-3 activity was determined by fluorimetric assay. Oil red O (ORO) staining was performed for fat detection. Apoptotic cells were identified by morphological criteria, immunohistochemistry for caspase-3, and terminal deoxynucleotidyl transferase-mediated 2'-deoxyuridine 5'-triphosphate nick-end labeling (TUNEL) assay. At 1 minute of reperfusion, the ischemic (Bax-/-) livers were characterized by statistically significantly lower liver enzyme levels and lower caspase-3 activity than the ischemic (Bax+/+) livers (P<0.05 for both). The reduction in postischemic apoptotic hepatic injury in the ischemic Bax-/- livers group was confirmed morphologically, by the significantly reduced microvesicular steatosis as determined by ORO staining, fewer apoptotic hepatocyte cells detected (P<0.05); immunohistochemically, by the significantly weaker activation of caspase-3 compared to the ischemic group (P<0.05); and by TUNEL assay (P<0.05). Similar levels of antiapoptotic Bcl-2 protein expression were detected in all 3 groups of ischemic livers on Western blots. Bax protein was not expressed in Bax-deficient livers and was detected in Bax+/+ normal livers. In the Bax+/- livers, levels of the damage markers were moderate. In conclusion, The better tolerance of Bax knockout livers to I/R injury suggests that the Bax gene may serve as a potential target for therapeutic intervention in hepatic I/R injury.