Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Biomed Res Int ; 2015: 417590, 2015.
Article in English | MEDLINE | ID: mdl-25861623

ABSTRACT

Liver ischemia-reperfusion (I-R) injury occurs during liver resection, liver transplantation, and hemorrhagic shock. The main mode of liver cell death after warm and/or cold liver I-R is necrosis, but other modes of cell death, as apoptosis and autophagy, are also involved. Autophagy is an intracellular self-digesting pathway responsible for removal of long-lived proteins, damaged organelles, and malformed proteins during biosynthesis by lysosomes. Autophagy is found in normal and diseased liver. Although depending on the type of ischemia, warm and/or cold, the dynamic process of liver I-R results mainly in adenosine triphosphate depletion and in production of reactive oxygen species (ROS), leads to both, a local ischemic insult and an acute inflammatory-mediated reperfusion injury, and results finally in cell death. This process can induce liver dysfunction and can increase patient morbidity and mortality after liver surgery and hemorrhagic shock. Whether autophagy protects from or promotes liver injury following warm and/or cold I-R remains to be elucidated. The present review aims to summarize the current knowledge in liver I-R injury focusing on both the beneficial and the detrimental effects of liver autophagy following warm and/or cold liver I-R.


Subject(s)
Autophagy/physiology , Liver/pathology , Reperfusion Injury/pathology , Animals , Humans , Liver Diseases/pathology
3.
J Transplant ; 2012: 164329, 2012.
Article in English | MEDLINE | ID: mdl-22530107

ABSTRACT

Ischemia-reperfusion (I-R) injury after liver transplantation (LT) induces intra- and/or extrahepatic nonanastomotic ischemic-type biliary lesions (ITBLs). Subsequent bile duct stricture is a significant cause of morbidity and even mortality in patients who underwent LT. Although the pathogenesis of ITBLs is multifactorial, there are three main interconnected mechanisms responsible for their formation: cold and warm I-R injury, injury induced by cytotoxic bile salts, and immunological-mediated injury. Cold and warm ischemic insult can induce direct injury to the cholangiocytes and/or damage to the arterioles of the peribiliary vascular plexus, which in turn leads to apoptosis and necrosis of the cholangiocytes. Liver grafts from suboptimal or extended-criteria donors are more susceptible to cold and warm I-R injury and develop more easily ITBLs than normal livers. This paper, focusing on liver I-R injury, reviews the risk factors and mechanisms leading to ITBLs following LT.

5.
Langenbecks Arch Surg ; 394(1): 123-31, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18679708

ABSTRACT

BACKGROUND: Phosphoregulation of signal transduction pathways is a complex series of reactions that may modulate the cellular response to ischemia-reperfusion (I-R). The aim of this study was to evaluate the effect of normothermic liver I/R-induced apoptosis on phosphorylation and activation of signal proteins in tyrosine kinase pathways. MATERIALS AND METHODS: In rats, a segmental normothermic ischemia of the liver was induced for 120 min. Liver apoptosis was determined using terminal deoxynucleotide-transferase-mediated deoxyuridine triphosphate nick end labeling assay, and activity of caspases-3 and -7 was determined by fluorescence. Liver tyrosine phosphorylation of proteins was examined by Western blot analysis. RESULTS: Normothermic I-R resulted in increased in vivo caspases-3 and -7 activity and in liver apoptosis. Shc tyrosine phosphorylation and activation of ERK1/2 were increased after reperfusion, while tyrosine phosphorylation of IRS-1 and activation of PKB/Akt were decreased. CONCLUSIONS: Normothermic liver I-R leads to increased apoptosis and to modifications in protein tyrosine phosphorylation pathways.


Subject(s)
Apoptosis/physiology , Insulin Receptor Substrate Proteins/metabolism , Liver/blood supply , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Reperfusion Injury/physiopathology , Signal Transduction/physiology , Animals , Caspase 3/metabolism , Caspase 7/metabolism , In Situ Nick-End Labeling , Liver/pathology , Male , Phosphorylation , Rats , Rats, Inbred Lew , Receptor, Insulin/metabolism
6.
J Invest Surg ; 20(2): 97-103, 2007.
Article in English | MEDLINE | ID: mdl-17454394

ABSTRACT

In hemorrhagic shock (HS) an increased thymic apoptosis (TA) was described. The aim of this study was to evaluate the effect of administration of the caspase inhibitor N-benzyloxy-carbonil-Val-Ala-Asp-fluoromethylketone (Z-VAD-FMK) during the resuscitation phase on TA, organ dysfunctions, and tumor necrosis factor (TNF)-alpha release in HS. Forty rats were randomly assigned to four groups: no HS/resuscitation (sham); HS/resuscitation with shed blood and normal saline (control); HS/resuscitation with shed blood and phosphate-buffered solution (PBS) (vehicle); and HS/resuscitation with shed blood and Z-VAD-FMK (inhibitor). Rats were subjected to HS by blood removal to a MAP of 35-40 mmHg. After a 1-h shock period, the animals were resuscitated according to the protocol. At 1 and 3 h after resuscitation, transaminases, creatinine, urea, lipase, TNF-alpha, and TA were evaluated. Our study showed that a nonlethal HS is early able to induce organ dysfunctions and increased TA. Administration of Z-VAD-FMK did not significantly decrease organ dysfunctions, while it induced a significant TNF-alpha release. TA was significantly reduced by Z-VAD-FMK after 1 h, but not after 3 h. Our results suggest that postinjury caspase inhibition does not attenuate organ dysfunctions, and also does not permanently reduce TA induced by HS and resuscitation in rats.


Subject(s)
Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Enzyme Inhibitors/pharmacology , Shock, Hemorrhagic/pathology , Thymus Gland/drug effects , Thymus Gland/pathology , Animals , Caspases/metabolism , Male , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/metabolism
7.
HPB (Oxford) ; 9(2): 112-9, 2007.
Article in English | MEDLINE | ID: mdl-18333125

ABSTRACT

BACKGROUND: Pentoxifylline (PTX) has been shown to reduce hepatic injury after normothermic ischemia and reperfusion (I-R) in rat liver. AIM: The aim of this study was to evaluate the effects of PTX on liver expression of tumor necrosis factor alpha (TNFalpha) mRNA following normothermic liver I-R. MATERIALS AND METHODS: A segmental normothermic ischemia of the liver was induced in male Lewis rats by occluding the blood vessels including the bile duct to the median and left lateral lobes for 90 min. At the end of ischemia the nonischemic liver lobes were resected. Rats were divided into three groups: group 1, control Ringer lactate administration; group 2, PTX treatment; group 3, sham-operated control rats. PTX (50 mg/kg) was injected intravenously 30 min before and 60 min after induction of ischemia. Survival rates were compared and the serum activities of TNFalpha, serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and lactate dehydrogenase (LDH) were measured. Histology of the liver was assessed 6 h after reperfusion. Liver TNFalpha mRNA was assessed by PCR amplification at 0, 60, 120, and 210 min after reperfusion. RESULTS: PTX treatment significantly increased 7 day survival (93.3%) compared with nontreated control rats (46.6%, p<0.007). The extent of liver necrosis and the release of liver enzymes were significantly decreased after PTX treatment. Serum activities of TNFalpha were significantly decreased and liver expression of TNFalpha mRNA was inhibited after PTX treatment. CONCLUSION: PTX protects the liver from ischemic injury and inhibits liver expression of TNFalpha mRNA.

8.
J Surg Res ; 125(1): 30-6, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15836847

ABSTRACT

BACKGROUND: The aim of this study was to evaluate the role of the pro-apoptotic molecule Fas Ligand (FasL) in 120 min normothermic ischemia-reperfusion (I-R) induced apoptosis in rat liver treated or not with Z-Asp-cmk caspase inhibitor. MATERIALS AND METHODS: Rats were divided into two groups: group 1, control, PBS administration; group 2, Z-Asp-cmk treatment. Z-Asp-cmk was injected intravenously, 2 min before induction of 120 min of normothermic liver ischemia. Immunohistochemical detection of apoptotic liver cells was carried out using the TUNEL method. Fas and FasL expression were measured by qualitative reverse transcription polymerase chain reaction (RT-PCR), Northern and western blot, and by immunofluorescence labeling, in ischemic and non-ischemic liver lobes at different times after reperfusion. RESULTS: FasL mRNA and protein expression were increased in ischemic liver, while Fas receptor mRNA levels remained unchanged. Pre-treatment of rats with Z-Asp-cmk caspase inhibitor reduced liver apoptosis, but did not modify FasL mRNA levels. CONCLUSIONS: These results suggest that the pro-apoptotic molecule FasL is involved in the induction of liver apoptosis following I-R.


Subject(s)
Liver/blood supply , Membrane Glycoproteins/genetics , RNA, Messenger/analysis , Reperfusion , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis , Caspases/metabolism , Fas Ligand Protein , In Situ Nick-End Labeling , Male , Rats , Rats, Inbred Lew
9.
Eur J Pharmacol ; 473(2-3): 177-84, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12892836

ABSTRACT

The aim of this study was to demonstrate that tacrolimus (FK506) has a hepatoprotective effect by reducing ischemia-reperfusion-induced apoptosis and necrosis, both of which lead to post-surgical liver dysfunction. An ischemia-reperfusion model and primary cultured rat hepatocytes subjected to hypoxic and reoxygenation phases, mimicking the surgical process, were used. c-Jun N-terminal kinase 1/stress-activated protein kinase 1 (JNK1/SAPK1) activation leads to caspase 3 activation, a trigger of apoptosis. The activation status of JNK1/SAPK1 was evaluated by immunoprecipitation or Western-blotting experiments. Apoptosis was assessed by measuring caspase activation and by TUNEL (terminal deoxynucleotidyltransferase-mediated deoxyuridine triphosphate-biotin nick-end labeling) reaction. Necrosis was assessed histologically. Tacrolimus improved the survival rate of rats subjected to ischemia-reperfusion. After FK506 pretreatment, the liver necrosis rate was reduced, and ischemia-reperfusion-induced JNK1/SAPK1 activation and apoptosis were significantly decreased. In hypoxia-reoxygenation-subjected hepatocytes, tacrolimus reduced JNK1/SAPK1 and caspase 3 activation. In the liver, tacrolimus prevented ischemia-reperfusion-induced apoptosis and necrosis.


Subject(s)
Apoptosis/drug effects , Immunosuppressive Agents/pharmacology , Ischemia/drug therapy , Liver/drug effects , Reperfusion Injury/prevention & control , Tacrolimus/pharmacology , Animals , Blotting, Western , Caspases/metabolism , Cells, Cultured , Hepatocytes/drug effects , Hepatocytes/enzymology , Hepatocytes/metabolism , In Situ Nick-End Labeling , Ischemia/metabolism , Ischemia/pathology , JNK Mitogen-Activated Protein Kinases , Liver/blood supply , Liver/pathology , Male , Mitogen-Activated Protein Kinases/metabolism , Necrosis , Rats , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
10.
Dig Dis Sci ; 48(6): 1147-58, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12822878

ABSTRACT

Previous study pointed to an important role of adrenals and glucocorticoids in the trophic status of the adult small intestine mucosa, with possible implications during stress events. Small intestine morphological and biochemical consequences of 10-day bilateral adrenalectomy and also sham-related laparotomy were determined in 23-month-old Sprague-Dawley rats. As described in young rats, adrenalectomy in old rats leads to partial atrophy and disorganization of the proximal small intestine epithelium, with an increase in the number of Paneth cells and reduced crypt cell proliferation. We also observed a decrease of goblet cell number and a reduction of all enzyme activities including disaccharidases, in contrast with the specific induced response shown in young rats. A number of marked biochemical effects have also been noted in aged rats subjected to solely laparotomy, suggesting age-related adaptation impairments. In conclusion, adrenalectomy modified the differentiation processes of the small intestinal mucosa in both young and aged rats, and some parameters underlined that the lack of corticoid-mediated adaptive process are exacerbated by cumulative surgical stress (event) and aging.


Subject(s)
Adaptation, Physiological/physiology , Adrenalectomy , Aging/physiology , Glucocorticoids/physiology , Intestine, Small/growth & development , Animals , Body Weight , Cell Division , Eating , Intestinal Mucosa/pathology , Intestine, Small/physiopathology , Male , Rats , Rats, Sprague-Dawley , Stress, Physiological/physiopathology
11.
Pharmacology ; 65(2): 87-95, 2002 May.
Article in English | MEDLINE | ID: mdl-11937779

ABSTRACT

Interruption of hepatic blood flow is necessary in surgery, but the liver is sensitive to ischemia and reperfusion. Hypoxia induces an increase in intracellular calcium concentration. In previous studies, we have shown that hypoxia-reoxygenation (H/R) increased calcium influx and induced JNK(1)/SAPK(1) activation which was involved in the triggering of apoptosis. The aim of this study was to demonstrate that diltiazem, a calcium inhibitor, reduced JNK(1)/SAPK(1) activation and consequently could decrease H/R-induced apoptosis. Experiments were performed, in the presence of diltiazem, on primary cultured rat hepatocytes, subjected to warm H/R phases and in a liver ischemia-reperfusion model. The activation status of JNK(1)/SAPK(1) was evaluated by immunoprecipitation and immunohistolocalisation experiments, while apoptosis was assessed by measuring caspase activity and by TUNEL labeling. Diltiazem inhibited H/R-induced JNK(1)/SAPK(1) activation and decreased apoptosis. It could be used to improve postoperative liver function.


Subject(s)
Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Diltiazem/pharmacology , Hepatocytes/drug effects , Hypoxia/physiopathology , Oxygen/pharmacology , Animals , Apoptosis/physiology , Calcium/metabolism , Cells, Cultured , Enzyme Activation/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Hot Temperature , Liver/blood supply , Liver/drug effects , Liver/enzymology , Male , Mitogen-Activated Protein Kinase 8 , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Rats , Rats, Wistar , Reperfusion Injury/physiopathology
12.
FASEB J ; 16(1): 93-5, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11709491

ABSTRACT

Hepatic ischemia occurs in liver transplantation, hemodynamic or cardiogenic shock, and liver resection associated with trauma or tumor. Ischemia/reperfusion (I/R) injury results in microcirculation failure followed by apoptosis and necrosis. Matrix metalloproteinases (MMPs) are involved in many physiological and pathological processes, but their expression and function during liver I/R remains poorly documented. In this study, we evaluated the expression of nine MMPs and their natural inhibitors, tissue inhibitors of MMPs (TIMPs), in a rat model of liver I/R. Analysis of MMP and TIMP expression show that although most of these genes are not constitutively expressed in the normal liver, they are induced in a specific time-dependent manner following I/R. Stromelysin-1, gelatinase B, and collagenase-3 are induced during the early phase of acute liver injury associated with inflammation and increased necrosis/apoptosis, whereas gelatinase A, membrane type-MMP, stromelysin-3, metalloelastase, TIMP-1, and TIMP-2 are essentially detectable during the recovery phase of liver injury corresponding to hepatocyte regeneration. This observation suggested that MMPs and TIMPs could play both deleterious and beneficial roles following I/R. We thus tested the effect of a specific phosphinic MMP inhibitor on acute liver I/R injury. Inhibition of MMP activity was shown to significantly decrease liver injury in ischemic/reperfused liver tissue as assessed by histological studies and serum hepatic enzyme levels. We therefore propose that MMP inhibitors may be of clinical relevance in liver-associated ischemic diseases or after liver transplantation.


Subject(s)
Liver Diseases/prevention & control , Matrix Metalloproteinase Inhibitors , Reperfusion Injury/prevention & control , Animals , Apoptosis/drug effects , Liver/drug effects , Liver/enzymology , Liver Diseases/enzymology , Liver Diseases/pathology , Liver Regeneration , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Necrosis , Phosphinic Acids/pharmacology , RNA, Messenger/biosynthesis , Rats , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Temperature , Tissue Inhibitor of Metalloproteinases/biosynthesis , Tissue Inhibitor of Metalloproteinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...