Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Redox Biol ; 9: 188-197, 2016 10.
Article in English | MEDLINE | ID: mdl-27566282

ABSTRACT

BACKGROUND: Mitochondrial dysfunction and bioenergetic stress play an important role in the etiology of alcoholic liver disease. Previous studies from our laboratory show that the primary methyl donor S-Adenosylmethionine (SAM) minimizes alcohol-induced disruptions in several mitochondrial functions in the liver. Herein, we expand on these earlier observations to determine whether the beneficial actions of SAM against alcohol toxicity extend to changes in the responsiveness of mitochondrial respiration to inhibition by nitric oxide (NO), induction of the mitochondrial permeability transition (MPT) pore, and the hypoxic state of the liver. METHODS: For this, male Sprague-Dawley rats were pair-fed control and alcohol-containing liquid diets with and without SAM for 5 weeks and liver hypoxia, mitochondrial respiration, MPT pore induction, and NO-dependent control of respiration were examined. RESULTS: Chronic alcohol feeding significantly enhanced liver hypoxia, whereas SAM supplementation attenuated hypoxia in livers of alcohol-fed rats. SAM supplementation prevented alcohol-mediated decreases in mitochondrial state 3 respiration and cytochrome c oxidase activity. Mitochondria isolated from livers of alcohol-fed rats were more sensitive to calcium-mediated MPT pore induction (i.e., mitochondrial swelling) than mitochondria from pair-fed controls, whereas SAM treatment normalized sensitivity for calcium-induced swelling in mitochondria from alcohol-fed rats. Liver mitochondria from alcohol-fed rats showed increased sensitivity to NO-dependent inhibition of respiration compared with pair-fed controls. In contrast, mitochondria isolated from the livers of SAM treated alcohol-fed rats showed no change in the sensitivity to NO-mediated inhibition of respiration. CONCLUSION: Collectively, these findings indicate that the hepato-protective effects of SAM against alcohol toxicity are mediated, in part, through a mitochondrial mechanism involving preservation of key mitochondrial bioenergetic parameters and the attenuation of hypoxic stress.


Subject(s)
Fatty Liver, Alcoholic/metabolism , Hypoxia/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Organelle Biogenesis , S-Adenosylmethionine/metabolism , Animals , Biomarkers , Cell Respiration , Disease Models, Animal , Electron Transport Complex I/metabolism , Electron Transport Complex IV/metabolism , Ethanol/adverse effects , Ethanol/metabolism , Fatty Liver, Alcoholic/pathology , Liver/drug effects , Liver/pathology , Mitochondria, Liver/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Nitric Oxide/metabolism , Rats , Reactive Oxygen Species/metabolism , S-Adenosylmethionine/pharmacology
2.
Nat Commun ; 6: 7792, 2015 Aug 05.
Article in English | MEDLINE | ID: mdl-26242746

ABSTRACT

Macrophages are an essential component of the immune response to ischaemic injury and play an important role in promoting inflammation and its resolution, which is necessary for tissue repair. The type I transmembrane glycoprotein CD163 is exclusively expressed on macrophages, where it acts as a receptor for haemoglobin:haptoglobin complexes. An extracellular portion of CD163 circulates in the blood as a soluble protein, for which no physiological function has so far been described. Here we show that during ischaemia, soluble CD163 functions as a decoy receptor for TWEAK, a secreted pro-inflammatory cytokine of the tumour necrosis factor family, to regulate TWEAK-induced activation of canonical nuclear factor-κB (NF-κB) and Notch signalling necessary for myogenic progenitor cell proliferation. Mice with deletion of CD163 have transiently elevated levels of TWEAK, which stimulate muscle satellite cell proliferation and tissue regeneration in their ischaemic and non-ischaemic limbs. These results reveal a role for soluble CD163 in regulating muscle regeneration after ischaemic injury.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Macrophages/physiology , Muscle, Skeletal/physiology , Receptors, Cell Surface/metabolism , Regeneration , Tumor Necrosis Factors/metabolism , Animals , Cytokine TWEAK , Male , Mice, Knockout , NF-kappa B/metabolism , Random Allocation , Receptors, Notch/metabolism , Reperfusion Injury
3.
Biochem J ; 461(2): 223-32, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24758559

ABSTRACT

NAFLD (non-alcoholic fatty liver disease) involves significant changes in liver metabolism characterized by oxidative stress, lipid accumulation and fibrogenesis. Mitochondrial dysfunction and bioenergetic defects also contribute to NAFLD. In the present study, we examined whether differences in mtDNA influence NAFLD. To determine the role of mitochondrial and nuclear genomes in NAFLD, MNX (mitochondrial-nuclear exchange) mice were fed an atherogenic diet. MNX mice have mtDNA from C57BL/6J mice on a C3H/HeN nuclear background and vice versa. Results from MNX mice were compared with wild-type C57BL/6J and C3H/HeN mice fed a control or atherogenic diet. Mice with the C57BL/6J nuclear genome developed more macrosteatosis, inflammation and fibrosis compared with mice containing the C3H/HeN nuclear genome when fed the atherogenic diet. These changes were associated with parallel alterations in inflammation and fibrosis gene expression in wild-type mice, with intermediate responses in MNX mice. Mice with the C57BL/6J nuclear genome had increased State 4 respiration, whereas MNX mice had decreased State 3 respiration and RCR (respiratory control ratio) when fed the atherogenic diet. Complex IV activity and most mitochondrial biogenesis genes were increased in mice with the C57BL/6J nuclear or mitochondrial genome, or both fed the atherogenic diet. These results reveal new interactions between mitochondrial and nuclear genomes and support the concept that mtDNA influences mitochondrial function and metabolic pathways implicated in NAFLD.


Subject(s)
Cell Nucleus/metabolism , Fatty Liver/genetics , Genome, Mitochondrial , Hepatocytes/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Animals , Cell Nucleus/pathology , Diet, Atherogenic/adverse effects , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/pathology , Fibrosis , Gene Expression , Gene Expression Profiling , Hepatocytes/pathology , Inflammation/etiology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Liver/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease , Oxidative Phosphorylation , Severity of Illness Index
4.
Proc Natl Acad Sci U S A ; 111(8): 3182-7, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516168

ABSTRACT

Previous studies have demonstrated that hydrogen sulfide (H2S) protects against multiple cardiovascular disease states in a similar manner as nitric oxide (NO). H2S therapy also has been shown to augment NO bioavailability and signaling. The purpose of this study was to investigate the impact of H2S deficiency on endothelial NO synthase (eNOS) function, NO production, and ischemia/reperfusion (I/R) injury. We found that mice lacking the H2S-producing enzyme cystathionine γ-lyase (CSE) exhibit elevated oxidative stress, dysfunctional eNOS, diminished NO levels, and exacerbated myocardial and hepatic I/R injury. In CSE KO mice, acute H2S therapy restored eNOS function and NO bioavailability and attenuated I/R injury. In addition, we found that H2S therapy fails to protect against I/R in eNOS phosphomutant mice (S1179A). Our results suggest that H2S-mediated cytoprotective signaling in the setting of I/R injury is dependent in large part on eNOS activation and NO generation.


Subject(s)
Cytoprotection/physiology , Hydrogen Sulfide/metabolism , Myocardial Reperfusion Injury/drug therapy , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Signal Transduction/physiology , Alanine Transaminase/blood , Analysis of Variance , Animals , Aspartate Aminotransferases/blood , Blotting, Western , Chromatography, High Pressure Liquid , Cystathionine gamma-Lyase/genetics , Cytoprotection/drug effects , Hydrogen Sulfide/pharmacology , Immunohistochemistry , Mice , Mice, Knockout , Mitochondria/physiology , Myocardial Reperfusion Injury/metabolism , Oxidative Stress/physiology , Oxygen Consumption/physiology , Troponin I/metabolism
5.
Am J Physiol Gastrointest Liver Physiol ; 306(4): G265-77, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24356880

ABSTRACT

Chronic ethanol consumption increases sensitivity of the mitochondrial permeability transition (MPT) pore induction in liver. Ca(2+) promotes MPT pore opening, and genetic ablation of cyclophilin D (CypD) increases the Ca(2+) threshold for the MPT. We used wild-type (WT) and CypD-null (CypD(-/-)) mice fed a control or an ethanol-containing diet to investigate the role of the MPT in ethanol-mediated liver injury. Ca(2+)-mediated induction of the MPT and mitochondrial respiration were measured in isolated liver mitochondria. Steatosis was present in WT and CypD(-/-) mice fed ethanol and accompanied by increased terminal deoxynucleotidyl transferase dUTP-mediated nick-end label-positive nuclei. Autophagy was increased in ethanol-fed WT mice compared with ethanol-fed CypD(-/-) mice, as reflected by an increase in the ratio of microtubule protein 1 light chain 3B II to microtubule protein 1 light chain 3B I. Higher levels of p62 were measured in CypD(-/-) than WT mice. Ethanol decreased mitochondrial respiratory control ratios and select complex activities in WT and CypD(-/-) mice. Ethanol also increased CypD protein in liver of WT mice. Mitochondria from control- and ethanol-fed WT mice were more sensitive to Ca(2+)-mediated MPT pore induction than mitochondria from their CypD(-/-) counterparts. Mitochondria from ethanol-fed CypD(-/-) mice were also more sensitive to Ca(2+)-induced swelling than mitochondria from control-fed CypD(-/-) mice but were less sensitive than mitochondria from ethanol-fed WT mice. In summary, CypD deficiency was associated with impaired autophagy and did not prevent ethanol-mediated steatosis. Furthermore, increased MPT sensitivity was observed in mitochondria from ethanol-fed WT and CypD(-/-) mice. We conclude that chronic ethanol consumption likely lowers the threshold for CypD-regulated and -independent characteristics of the ethanol-mediated MPT pore in liver mitochondria.


Subject(s)
Ethanol , Liver Diseases, Alcoholic/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Autophagy , Calcium Signaling , Cell Respiration , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/genetics , Disease Models, Animal , Fatty Liver, Alcoholic/etiology , Fatty Liver, Alcoholic/metabolism , Genotype , Liver/pathology , Liver Diseases, Alcoholic/etiology , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Mitochondria, Liver/pathology , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling , Phenotype , Time Factors
6.
Circulation ; 127(10): 1116-27, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23393010

ABSTRACT

BACKGROUND: Cystathionine γ-lyase (CSE) produces H2S via enzymatic conversion of L-cysteine and plays a critical role in cardiovascular homeostasis. We investigated the effects of genetic modulation of CSE and exogenous H2S therapy in the setting of pressure overload-induced heart failure. METHODS AND RESULTS: Transverse aortic constriction was performed in wild-type, CSE knockout, and cardiac-specific CSE transgenic mice. In addition, C57BL/6J or CSE knockout mice received a novel H2S donor (SG-1002). Mice were followed up for 12 weeks with echocardiography. We observed a >60% reduction in myocardial and circulating H2S levels after transverse aortic constriction. CSE knockout mice exhibited significantly greater cardiac dilatation and dysfunction than wild-type mice after transverse aortic constriction, and cardiac-specific CSE transgenic mice maintained cardiac structure and function after transverse aortic constriction. H2S therapy with SG-1002 resulted in cardioprotection during transverse aortic constriction via upregulation of the vascular endothelial growth factor-Akt-endothelial nitric oxide synthase-nitric oxide-cGMP pathway with preserved mitochondrial function, attenuated oxidative stress, and increased myocardial vascular density. CONCLUSIONS: Our results demonstrate that H2S levels are decreased in mice in the setting of heart failure. Moreover, CSE plays a critical role in the preservation of cardiac function in heart failure, and oral H2S therapy prevents the transition from compensated to decompensated heart failure in part via upregulation of endothelial nitric oxide synthase and increased nitric oxide bioavailability.


Subject(s)
Cardiotonic Agents/therapeutic use , Heart Failure/drug therapy , Heart Failure/enzymology , Hydrogen Sulfide/therapeutic use , Nitric Oxide Synthase Type III/biosynthesis , Up-Regulation/drug effects , Animals , Cardiotonic Agents/administration & dosage , Cystathionine gamma-Lyase/deficiency , Cystathionine gamma-Lyase/genetics , Heart Failure/physiopathology , Hydrogen Sulfide/administration & dosage , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type III/physiology , Up-Regulation/physiology
7.
Arterioscler Thromb Vasc Biol ; 32(8): 1865-74, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22652602

ABSTRACT

OBJECTIVE: ß(2)-adrenoreceptor activation has been shown to protect cardiac myocytes from cell death. We hypothesized that acute ß(2)-adrenoreceptor stimulation, using arformoterol (ARF), would attenuate myocardial ischemia/reperfusion (R) injury via NO synthase activation and cause a subsequent increase in NO bioavailability. METHODS AND RESULTS: Male C57BL/6J and endothelial NO synthase (eNOS) knockout mice were subjected to 45 minutes of myocardial ischemia and 24 hours of R. ARF or vehicle was administered 5 minutes before R. Serum troponin-I was measured, and infarct size per area-at-risk was evaluated at 24 hours of R. Echocardiography was performed at baseline and 2 weeks after R. Myocardial cAMP, protein kinase A, eNOS/Akt phosphorylation status, and NO metabolite levels were assayed. ARF (1 µg/kg) reduced infarct size per area-at-risk by 53.1% (P<0.001 versus vehicle) and significantly reduced troponin-I levels (P<0.001 versus vehicle). Ejection fraction was significantly preserved in ARF-treated hearts compared with vehicle hearts at 2 weeks of R. Serum cAMP and nuclear protein kinase A C-α increased 5 and 15 minutes after ARF injection, respectively (P<0.01). ARF increased Akt phosphorylation at Thr(308) (P<0.001) and Ser(473) (P<0.01), and eNOS phosphorylation at Ser(1177) (P<0.01). ARF treatment increased heart nitrosothiol levels (P<0.001) at 15 min after injection. ARF failed to reduce infarct size in eNOS(-/-) mice. CONCLUSIONS: Our results indicate that ß(2)-adrenoreceptor stimulation activates cAMP, protein kinase A, Akt, and eNOS and augments NO bioavailability. Activation of this prosurvival signaling pathway attenuates myocardial cell death and preserves cardiac function after ischemia/reperfusion.


Subject(s)
Adrenergic beta-2 Receptor Agonists/therapeutic use , Ethanolamines/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocytes, Cardiac/pathology , Animals , Cell Death/drug effects , Cyclic AMP-Dependent Protein Kinases/physiology , Formoterol Fumarate , Ischemic Preconditioning, Myocardial , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Proteins c-akt/metabolism
8.
Am J Physiol Heart Circ Physiol ; 302(11): H2410-8, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22467307

ABSTRACT

Diallyl trisulfide (DATS), a polysulfide constituent found in garlic oil, is capable of the release of hydrogen sulfide (H(2)S). H(2)S is a known cardioprotective agent that protects the heart via antioxidant, antiapoptotic, anti-inflammatory, and mitochondrial actions. Here, we investigated DATS as a stable donor of H(2)S during myocardial ischemia-reperfusion (MI/R) injury in vivo. We investigated endogenous H(2)S levels, infarct size, postischemic left ventricular function, mitochondrial respiration and coupling, endothelial nitric oxide (NO) synthase (eNOS) activation, and nuclear E2-related factor (Nrf2) translocation after DATS treatment. Mice were anesthetized and subjected to a surgical model of MI/R injury with and without DATS treatment (200 µg/kg). Both circulating and myocardial H(2)S levels were determined using chemiluminescent gas chromatography. Infarct size was measured after 45 min of ischemia and 24 h of reperfusion. Troponin I release was measured at 2, 4, and 24 h after reperfusion. Cardiac function was measured at baseline and 72 h after reperfusion by echocardiography. Cardiac mitochondria were isolated after MI/R, and mitochondrial respiration was investigated. NO metabolites, eNOS phosphorylation, and Nrf2 translocation were determined 30 min and 2 h after DATS administration. Myocardial H(2)S levels markedly decreased after I/R injury but were rescued by DATS treatment (P < 0.05). DATS administration significantly reduced infarct size per area at risk and per left ventricular area compared with control (P < 0.001) as well as circulating troponin I levels at 4 and 24 h (P < 0.05). Myocardial contractile function was significantly better in DATS-treated hearts compared with vehicle treatment (P < 0.05) 72 h after reperfusion. DATS reduced mitochondrial respiration in a concentration-dependent manner and significantly improved mitochondrial coupling after reperfusion (P < 0.01). DATS activated eNOS (P < 0.05) and increased NO metabolites (P < 0.05). DATS did not appear to significantly induce the Nrf2 pathway. Taken together, these data suggest that DATS is a donor of H(2)S that can be used as a cardioprotective agent to treat MI/R injury.


Subject(s)
Allyl Compounds/therapeutic use , Hydrogen Sulfide/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Nitric Oxide/metabolism , Sulfides/therapeutic use , Allyl Compounds/pharmacology , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/physiology , Models, Animal , Myocardium/metabolism , Sulfides/pharmacology , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
9.
Int J Hepatol ; 2012: 962183, 2012.
Article in English | MEDLINE | ID: mdl-22187660

ABSTRACT

Introduction. Mitochondrial damage and disruption in oxidative phosphorylation contributes to the pathogenesis of alcoholic liver injury. Herein, we tested the hypothesis that the hepatoprotective actions of betaine against alcoholic liver injury occur at the level of the mitochondrial proteome. Methods. Male Wister rats were pair-fed control or ethanol-containing liquid diets supplemented with or without betaine (10 mg/mL) for 4-5 wks. Liver was examined for triglyceride accumulation, levels of methionine cycle metabolites, and alterations in mitochondrial proteins. Results. Chronic ethanol ingestion resulted in triglyceride accumulation which was attenuated in the ethanol plus betaine group. Blue native gel electrophoresis (BN-PAGE) revealed significant decreases in the content of the intact oxidative phosphorylation complexes in mitochondria from ethanol-fed animals. The alcohol-dependent loss in many of the low molecular weight oxidative phosphorylation proteins was prevented by betaine supplementation. This protection by betaine was associated with normalization of SAM : S-adenosylhomocysteine (SAH) ratios and the attenuation of the ethanol-induced increase in inducible nitric oxide synthase and nitric oxide generation in the liver. Discussion/Conclusion. In summary, betaine attenuates alcoholic steatosis and alterations to the oxidative phosphorylation system. Therefore, preservation of mitochondrial function may be another key molecular mechanism responsible for betaine hepatoprotection.

11.
Exp Physiol ; 96(9): 840-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21666033

ABSTRACT

Hydrogen sulfide (H(2)S) has been known as a highly toxic gas for several centuries. There have been considerable advances made in the H(2)S field regarding its physiological role; however, there is much more work that needs to be done. The biosynthesis of H(2)S has been attributed to three endogenous enzymes: cystathionine ß-synthase (CBS), cystathionine γ-lyase (CGL or CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST). These enzymes require further investigation to more fully elucidate the cellular expression profile, regulation and precise role of these critical enzymes in the production of H(2)S. In recent years, H(2)S has been demonstrated to have cytoprotective effects in multiple organ systems. In particular, it has been demonstrated that the administration of H(2)S either prior to ischaemia or at reperfusion significantly ameliorates myocardial and hepatic ischaemia-reperfusion injury. Therefore, this review focuses on the cardioprotective and hepatoprotective role of H(2)S. In addition, the review provides a summary of several known molecular targets of H(2)S protection.


Subject(s)
Cytoprotection , Hydrogen Sulfide/therapeutic use , Reperfusion Injury/prevention & control , Cystathionine beta-Synthase/metabolism , Cystathionine gamma-Lyase/metabolism , Humans , Hydrogen Sulfide/metabolism , Liver/blood supply , Liver/drug effects , Mitochondria/drug effects , Myocardial Reperfusion Injury/prevention & control , Potassium Channels/drug effects , Protein Processing, Post-Translational , Signal Transduction/drug effects , Sulfurtransferases/metabolism
12.
Antioxid Redox Signal ; 15(2): 447-59, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-20919931

ABSTRACT

Obesity-related pathologies, such as nonalcoholic fatty liver disease, are linked to mitochondrial dysfunction and nitric oxide (NO) deficiency. Herein, we tested the hypothesis that a high-fat diet (HFD) modifies the liver mitochondrial proteome and alters proteins involved in NO metabolism, namely arginase 1 and endothelial NO synthase. Male C57BL/6 mice were fed a control or HFD and liver mitochondria were isolated for proteomics and reactive oxygen species measurements. Steatosis and hepatocyte ballooning were present in livers of HFD mice, with no pathology observed in the controls. HFD mice had increased serum glucose and decreased adiponectin. Mitochondrial reactive oxygen species was increased after 8 weeks in the HFD mice, but decreased at 16 weeks compared with the control, which was accompanied by increased uncoupling protein 2. Using proteomics, 22 proteins were altered as a consequence of the HFD. This cohort consists of oxidative phosphorylation, lipid metabolism, sulfur amino acid metabolism, and chaperone proteins. We observed a HFD-dependent increase in arginase 1 and decrease in activated endothelial NO synthase. Serum and liver nitrate + nitrite were decreased by HFD. In summary, these data demonstrate that a HFD causes steatosis, alters NO metabolism, and modifies the liver mitochondrial proteome; thus, NO may play an important role in the processes responsible for nonalcoholic fatty liver disease.


Subject(s)
Dietary Fats/administration & dosage , Fatty Liver/etiology , Mitochondria, Liver/metabolism , Nitric Oxide/pharmacokinetics , Proteome , Animals , Biological Availability , Body Weight , Electrophoresis, Polyacrylamide Gel , Isoelectric Focusing , Male , Mice , Reactive Oxygen Species/metabolism
13.
Am J Physiol Gastrointest Liver Physiol ; 299(4): G954-66, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20651005

ABSTRACT

Chronic ethanol consumption increases mitochondrial oxidative stress and sensitivity to form the mitochondrial permeability transition pore (MPTP). The mechanism responsible for increased MPTP sensitivity in ethanol-exposed mitochondria and its relation to mitochondrial Ca(2+) handling is unknown. Herein, we investigated whether increased sensitivity to MPTP induction in liver mitochondria from ethanol-fed rats compared with controls is related to an ethanol-dependent change in mitochondrial Ca(2+) accumulation. Liver mitochondria were isolated from control and ethanol-fed rats, and Ca(2+)-mediated induction of the MPTP and mitochondrial Ca(2+) retention capacity were measured. Levels of proposed MPTP proteins as well as select pro- and antiapoptotic proteins were measured along with gene expression. We observed increased steatosis and TUNEL-stained nuclei in liver of ethanol-fed rats compared with controls. Liver mitochondria from ethanol-fed rats had increased levels of proapoptotic Bax protein and reduced Ca(2+) retention capacity compared with control mitochondria. We observed increased cyclophilin D (Cyp D) gene expression in liver and protein in mitochondria from ethanol-fed animals compared with controls, whereas there was no change in the adenine nucleotide translocase and voltage-dependent anion channel. Together, these results suggest that enhanced sensitivity to Ca(2+)-mediated MPTP induction may be due, in part, to higher Cyp D levels in liver mitochondria from ethanol-fed rats. Therefore, therapeutic strategies aimed at normalizing Cyp D levels may be beneficial in preventing ethanol-dependent mitochondrial dysfunction and liver injury.


Subject(s)
Calcium/metabolism , Cyclophilins/metabolism , Ethanol/adverse effects , Liver/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Animals , Peptidyl-Prolyl Isomerase F , Gene Expression Regulation/drug effects , Liver/metabolism , Male , Mitochondria, Liver/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Mitochondrial Permeability Transition Pore , Permeability , Rats , Rats, Sprague-Dawley
14.
Am J Physiol Gastrointest Liver Physiol ; 298(5): G732-45, 2010 May.
Article in English | MEDLINE | ID: mdl-20150243

ABSTRACT

S-adenosylmethionine (SAM) minimizes alcohol hepatotoxicity; however, the molecular mechanisms responsible for SAM hepatoprotection remain unknown. Herein, we use proteomics to determine whether the hepatoprotective action of SAM against early-stage alcoholic liver disease is linked to alterations in the mitochondrial proteome. For this, male rats were fed control or ethanol-containing liquid diets +/- SAM and liver mitochondria were prepared for proteomic analysis. Two-dimensional isoelectric focusing (2D IEF/SDS-PAGE) and blue native gel electrophoresis (BN-PAGE) were used to determine changes in matrix and oxidative phosphorylation (OxPhos) proteins, respectively. SAM coadministration minimized alcohol-dependent inflammation and preserved mitochondrial respiration. SAM supplementation preserved liver SAM levels in ethanol-fed rats; however, mitochondrial SAM levels were increased by ethanol and SAM treatments. With use of 2D IEF/SDS-PAGE, 30 proteins showed significant changes in abundance in response to ethanol, SAM, or both. Classes of proteins affected by ethanol and SAM treatments were chaperones, beta oxidation proteins, sulfur metabolism proteins, and dehydrogenase enzymes involved in methionine, glycine, and choline metabolism. BN-PAGE revealed novel changes in the levels of 19 OxPhos proteins in response to ethanol, SAM, or both. Ethanol- and SAM-dependent alterations in the proteome were not linked to corresponding changes in gene expression. In conclusion, ethanol and SAM treatment led to multiple changes in the liver mitochondrial proteome. The protective effects of SAM against alcohol toxicity are mediated, in part, through maintenance of proteins involved in key mitochondrial energy conserving and biosynthetic pathways. This study demonstrates that SAM may be a promising candidate for treatment of alcoholic liver disease.


Subject(s)
Ethanol/pharmacology , Liver Diseases, Alcoholic/prevention & control , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Proteome/drug effects , S-Adenosylmethionine/pharmacology , Animals , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Male , Mitochondria, Liver/chemistry , Mitochondrial Proteins/analysis , Oxygen Consumption/drug effects , Proteomics , Rats , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Transcription, Genetic/drug effects
15.
Curr Protoc Toxicol ; Chapter 14: Unit14.8, 2010 May.
Article in English | MEDLINE | ID: mdl-23045017

ABSTRACT

Mitochondrial dysfunction from toxicants is recognized as a causative factor in the development of numerous liver diseases including steatohepatitis, cirrhosis, and cancer. Toxicant-mediated damage to mitochondria result in depressed ATP production, inability to maintain proper cellular calcium homeostasis, and increased reactive oxygen species production. These disruptions contribute to hepatocellular death and lead to liver pathology. Herein, we describe a series of basic and advanced methodologies that can be incorporated into research projects aimed to understand the role of mitochondrial dysfunction in toxicant-induced hepatotoxicity. Protocols are provided for isolation of liver mitochondria, assessment of respiratory function, measurement of mitochondrial calcium uptake, and reactive oxygen species production, as well as characterization of the mitochondrial protein thiol proteome using 2D gel electrophoresis. Data obtained from these methods can be integrated into a logical and mechanistic framework to advance understanding of the role of mitochondrial dysfunction in the pathogenesis of toxicant-induced liver diseases.


Subject(s)
Biological Assay/methods , Chemical and Drug Induced Liver Injury , Mitochondria, Liver/drug effects , Mitochondrial Proteins/drug effects , Toxicity Tests/methods , Animals , Liver/drug effects , Liver/metabolism , Mice , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Proteomics/methods , Sulfhydryl Compounds/metabolism
16.
Biochem J ; 417(1): 183-93, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18752470

ABSTRACT

NAFLD (non-alcoholic fatty liver disease), associated with obesity and the cardiometabolic syndrome, is an important medical problem affecting up to 20% of western populations. Evidence indicates that mitochondrial dysfunction plays a critical role in NAFLD initiation and progression to the more serious condition of NASH (non-alcoholic steatohepatitis). Herein we hypothesize that mitochondrial defects induced by exposure to a HFD (high fat diet) contribute to a hypoxic state in liver and this is associated with increased protein modification by RNS (reactive nitrogen species). To test this concept, C57BL/6 mice were pair-fed a control diet and HFD containing 35% and 71% total calories (1 cal approximately 4.184 J) from fat respectively, for 8 or 16 weeks and liver hypoxia, mitochondrial bioenergetics, NO (nitric oxide)-dependent control of respiration, and 3-NT (3-nitrotyrosine), a marker of protein modification by RNS, were examined. Feeding a HFD for 16 weeks induced NASH-like pathology accompanied by elevated triacylglycerols, increased CYP2E1 (cytochrome P450 2E1) and iNOS (inducible nitric oxide synthase) protein, and significantly enhanced hypoxia in the pericentral region of the liver. Mitochondria from the HFD group showed increased sensitivity to NO-dependent inhibition of respiration compared with controls. In addition, accumulation of 3-NT paralleled the hypoxia gradient in vivo and 3-NT levels were increased in mitochondrial proteins. Liver mitochondria from mice fed the HFD for 16 weeks exhibited depressed state 3 respiration, uncoupled respiration, cytochrome c oxidase activity, and mitochondrial membrane potential. These findings indicate that chronic exposure to a HFD negatively affects the bioenergetics of liver mitochondria and this probably contributes to hypoxic stress and deleterious NO-dependent modification of mitochondrial proteins.


Subject(s)
Dietary Fats/administration & dosage , Fatty Liver/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , Animals , Cell Respiration/drug effects , Cytochrome P-450 CYP2E1/metabolism , Fatty Liver/etiology , Fatty Liver/pathology , Hypoxia/metabolism , Liver/drug effects , Liver/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondria, Liver/pathology , Mitochondrial Proteins/metabolism , Nitric Oxide/pharmacology , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Tyrosine/analogs & derivatives , Tyrosine/metabolism
17.
Free Radic Biol Med ; 44(7): 1259-72, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18242193

ABSTRACT

Fatty liver disease associated with chronic alcohol consumption or obesity/type 2 diabetes has emerged as a serious public health problem. Steatosis, accumulation of triglyceride in hepatocytes, is now recognized as a critical "first-hit" in the pathogenesis of liver disease. It is proposed that steatosis "primes" the liver to progress to more severe liver pathologies when individuals are exposed to subsequent metabolic and/or environmental stressors or "second-hits." Genetic risk factors can also influence the susceptibility to and severity of fatty liver disease. Furthermore, oxidative stress, disrupted nitric oxide (NO) signaling, and mitochondrial dysfunction are proposed to be key molecular events that accelerate or worsen steatosis and initiate progression to steatohepatitis and fibrosis. This review article will discuss the following topics regarding the pathobiology and molecular mechanisms responsible for fatty liver disease: (1) the "two-hit" or "multi-hit" hypothesis, (2) the role of mitochondrial bioenergetic defects and oxidant stress, (3) the interplay between NO and mitochondria in fatty liver disease, (4) genetic risk factors and oxidative stress-responsive genes, and (5) the feasibility of antioxidants for treatment.


Subject(s)
Alcohols/pharmacology , Fatty Liver/chemically induced , Mitochondria/pathology , Oxidative Stress , Animals , Antioxidants/pharmacology , Disease Progression , Fatty Acids/metabolism , Fatty Liver/etiology , Fibrosis/pathology , Free Radicals , Hepatocytes/cytology , Humans , Models, Biological , Obesity , Proteomics/methods
18.
World J Gastroenterol ; 13(37): 4967-73, 2007 Oct 07.
Article in English | MEDLINE | ID: mdl-17854139

ABSTRACT

Mitochondrial dysfunction is known to be a contributing factor to a number of diseases including chronic alcohol induced liver injury. While there is a detailed understanding of the metabolic pathways and proteins of the liver mitochondrion, little is known regarding how changes in the mitochondrial proteome may contribute to the development of hepatic pathologies. Emerging evidence indicates that reactive oxygen and nitrogen species disrupt mitochondrial function through post-translational modifications to the mitochondrial proteome. Indeed, various new affinity labeling reagents are available to test the hypothesis that post-translational modification of proteins by reactive species contributes to mitochondrial dysfunction and alcoholic fatty liver disease. Specialized proteomic techniques are also now available, which allow for identification of defects in the assembly of multi-protein complexes in mitochondria and the resolution of the highly hydrophobic proteins of the inner membrane. In this review knowledge gained from the study of changes to the mitochondrial proteome in alcoholic hepatotoxicity will be described and placed into a mechanistic framework to increase understanding of the role of mitochondrial dysfunction in liver disease.


Subject(s)
Liver Diseases, Alcoholic/metabolism , Mitochondria, Liver/metabolism , Reactive Oxygen Species/metabolism , Alcohol Drinking/metabolism , Humans , Liver Diseases, Alcoholic/etiology , Oxidative Stress/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...