Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Aliment Pharmacol Ther ; 57(10): 1131-1142, 2023 05.
Article in English | MEDLINE | ID: mdl-36864659

ABSTRACT

BACKGROUND AND AIMS: Little is known about the extent of mitochondrial respiratory chain (MRC) activity dysfunction in patients with alcoholic hepatitis (AH). We aimed to assess the hepatic MRC activity in AH patients and its potential impact on the severity and prognosis of this life-threatening liver disease. METHODS: MRC complexes were measured in liver biopsies of 98 AH patients (non-severe, 17; severe, 81) and in 12 histologically normal livers (NL). Severity was assessed according to Maddrey's Index and MELD score. Corticosteroid response rate and cumulative mortality were also evaluated. RESULTS: The activity of the five MRC complexes was markedly decreased in the liver of AH patients compared with that of NL subjects, being significantly lower in patients with severe AH than in those with non-severe AH. There was a negative correlation between the activity of all MRC complexes and the severity of AH. Interestingly, only complex I and III activities showed a significant positive correlation with the corticosteroid response rate and a significant negative correlation with the mortality rate at all-time points studied. In a multivariate regression analysis, besides the MELD score and the corticosteroid response rate, complex I activity was significantly associated with 3-month mortality (OR = 6.03; p = 0.034) and complex III activity with 6-month mortality (OR = 4.70; p = 0.041) in AH patients. CONCLUSION: Our results indicate that MRC activity is markedly decreased in the liver of AH patients, and, particularly, the impairment of MRC complexes I and III activity appears to have a significant impact on the clinical outcomes of patients with AH.


Subject(s)
Hepatitis, Alcoholic , Humans , Electron Transport , Prognosis , Adrenal Cortex Hormones , Severity of Illness Index
2.
Rev. esp. podol ; 34(2): 109-112, 2023. tab, ilus
Article in Spanish | IBECS | ID: ibc-229380

ABSTRACT

El carcinoma basocelular es el proceso neoformativo maligno que afecta más frecuentemente a la piel. Pese a que el diagnóstico debe ser siempre mediante estudio histopatológico, la ecografía puede sugerir la sospecha de este tipo de tumor. Se presenta un caso clínico en el que se sospechó de esta patología debido a la imagen ecográfica, posteriormente confirmado mediante el estudio histopatológico de la lesión (AU)


Basal cell carcinoma is the malignant neoformative process that most frequently affects the skin. Although the diagnosis must always be made through a histopathological study, ultrasound may suggest suspicion of this type of tumor. A clinical case is presented in which this disorder was suspected due to ultrasound image, later confirmed by the histopathological study of the lesion (AU)


Subject(s)
Humans , Male , Aged , Carcinoma, Basal Cell/diagnostic imaging , Carcinoma, Basal Cell/pathology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology
3.
Clin Lung Cancer ; 21(1): 75-85, 2020 01.
Article in English | MEDLINE | ID: mdl-31562055

ABSTRACT

BACKGROUND: Immunotherapy is a promising cancer treatment, but surrogate biomarkers of clinical efficacy have not been fully validated. The aim of this work was to evaluate several biomarkers as predictors of response to nivolumab monotherapy in patients with non-small-cell lung cancer. PATIENTS AND METHODS: Blood samples was collected at baseline, at 2 months after treatment start, and at disease progression. Lactate dehydrogenase level (LDH), neutrophils, and leukocyte values were obtained from medical record. Interleukin (IL)-8, IL-11, and kynurenine/tryptophan levels were determined by enzyme-linked immunosorbent assay. Total protein was extracted from circulating CD8+ T cells, and BCL-2 interacting mediator of cell death (BIM) protein expression tested by western blotting. RESULTS: Baseline LDH levels were significantly higher in non-responder patients than in those who responded (P = .045). The increase in indoleamine 2,3 dioxygenase activity was related to progression of disease, mainly in patients who did not respond to nivolumab treatment (P = .001). Increased levels of circulating IL-8 were observed in initially responding patients at time of progression, and it was related to lower overall survival (hazard ratio, 7.49; P = .025). A highest expression of BIM in circulating CD8+ T cells could be related to clinical benefit. The Student t test and Mann-Whitney U test were used to compare groups for continuous variables. Time to events was estimated using the Kaplan-Meier method, and compared by the log-rank test. CONCLUSIONS: Changes in plasma LDH and IL-8, indoleamine 2,3 dioxygenase activity, and BIM expression in CD8+ T cells could be used to monitor and predict clinical benefit from nivolumab treatment in these patients.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/pathology , Immunotherapy/methods , Lung Neoplasms/pathology , Nivolumab/therapeutic use , Adult , Aged , Aged, 80 and over , Bcl-2-Like Protein 11/blood , CD8-Positive T-Lymphocytes/pathology , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Female , Humans , Hydro-Lyases/blood , Indoleamine-Pyrrole 2,3,-Dioxygenase/blood , Interleukin-8/metabolism , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Male , Middle Aged , Survival Rate , Treatment Outcome
4.
Cancer Chemother Pharmacol ; 85(3): 525-535, 2020 03.
Article in English | MEDLINE | ID: mdl-31832811

ABSTRACT

PURPOSE: Therapeutic options for cancer patients have increased in the last years, although drugs resistance problem remains unresolved. Genetic background in individual susceptibility to cancer treatment could influence the therapy responses. The aim of this study was to explore the feasibility of using blood 4 genes (AEG-1, BRCA-1, REV3L and TYMS) expression levels as a predictor of the efficacy of pemetrexed therapy in patients with advanced non-small cell lung cancer. METHODS: Sixteen patients from the Medical Oncology Department at "12 de Octubre" Hospital, were included in the study. Total mRNA was isolated from blood samples, and gene expression was analyzed by RT-qPCR. A panel of lung tumor cell lines were used in cell proliferation tests and siRNA-mediated silencing assays. RESULTS: Similarity between blood gene expression levels and protein expression in matched tumor tissue was observed in 54.54% (REV3L) and 81.81% (TYMS) of cases. Gene expression of REV3L and TYMS in blood correlated directly and inversely, respectively, with progression-free survival and overall survival in the patients from our cohort. In tumor cell lines, the knockdown of REV3L conferred resistance to pemetrexed treatment, and the TYMS silencing increased the pemetrexed sensitivity of tumor cells. CONCLUSIONS: The use of peripheral blood samples for expression quantification of interest genes is an affordable method with promising results in the evaluation of response to pemetrexed treatment. Therefore, expression levels of REV3L and TYMS genes might be used as predictive biomarkers in advanced NSCLC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , DNA-Binding Proteins/genetics , DNA-Directed DNA Polymerase/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Thymidylate Synthase/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA-Binding Proteins/blood , DNA-Directed DNA Polymerase/blood , Female , Gene Expression/drug effects , Gene Expression/genetics , Gene Silencing/drug effects , Humans , Male , Middle Aged , Organoplatinum Compounds/therapeutic use , Pemetrexed/therapeutic use , Progression-Free Survival , Prospective Studies , RNA, Messenger/blood , RNA, Messenger/genetics , Thymidylate Synthase/blood
5.
Redox Biol ; 26: 101263, 2019 09.
Article in English | MEDLINE | ID: mdl-31299613

ABSTRACT

Inflammation is typically associated with the development of fibrosis, cirrhosis and hepatocellular carcinoma. The key role of protein tyrosine phosphatase 1B (PTP1B) in inflammatory responses has focused this study in understanding its implication in liver fibrosis. Here we show that hepatic PTP1B mRNA expression increased after bile duct ligation (BDL), while BDL-induced liver fibrosis was markedly reduced in mice lacking Ptpn1 (PTP1B-/-) as assessed by decreased collagen deposition and α-smooth muscle actin (α-SMA) expression. PTP1B-/- mice also showed a significant increase in mRNA levels of key markers of monocytes recruitment (Cd68, Adgre1 and Ccl2) compared to their wild-type (PTP1B+/+) littermates at early stages of injury after BDL. Interestingly, the lack of PTP1B strongly increased the NADPH oxidase (NOX) subunits Nox1/Nox4 ratio and downregulated Cybb expression after BDL, revealing a pro-survival pattern of NADPH oxidase induction in response to liver injury. Chimeric mice generated by transplantation of PTP1B-/- bone marrow (BM) into irradiated PTP1B+/+ mice revealed similar hepatic expression profile of NOX subunits than PTP1B-/- mice while these animals did not show differences in infiltration of myeloid cells at 7 days post-BDL, suggesting that PTP1B deletion in other liver cells is necessary for boosting the early inflammatory response to the BDL. PTP1B-/- BM transplantation into PTP1B+/+ mice also led to a blockade of TGF-ß and α-SMA induction after BDL. In vitro experiments demonstrated that deficiency of PTP1B in hepatocytes protects against bile acid-induced apoptosis and abrogates hepatic stellate cells (HSC) activation, an effect ameliorated by NOX1 inhibition. In conclusion, our results have revealed that the lack of PTP1B switches NOX expression pattern in response to liver injury after BDL and reduces HSC activation and liver fibrosis.


Subject(s)
Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , NADPH Oxidases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Animals , Apoptosis/genetics , Bile Acids and Salts/metabolism , Biomarkers , Cell Line , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Female , Gene Expression , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Immunohistochemistry , Kupffer Cells/metabolism , Liver Cirrhosis/pathology , Male , Mice , NADPH Oxidases/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/metabolism
6.
Int J Biometeorol ; 63(9): 1171-1180, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31227887

ABSTRACT

Spa mineral waters are used for the treatment of chronic diseases' symptoms. Anti-inflammatory, analgesic, anti-ageing and tissue repair effects have been attributed to them. This work seeks to improve knowledge about the effect of spa mineral waters on human cells. For this, human lung fibroblasts were treated with mineral waters from Ledesma, Paracuellos and Archena spas, three Spanish health resorts with different water chemical composition. A significant increase of cell proliferation together with an enhanced antioxidant capacity (reactive oxygen and nitrogen species, glutathione levels and superoxide dismutase activity) in mineral water-treated fibroblasts compared to control fibroblasts was observed. Moreover, cytokine profiling revealed an increase in the release of MIF, IL-6, CL-1, CCL-5 and ICAM-1, which are described as mediators in proliferation, wound healing and cell migration processes. In conclusion, our results could be in line with the effects attributed to spa mineral waters in wound healing strategies and oxidative damage protection.


Subject(s)
Antioxidants , Mineral Waters , Cell Line , Cell Proliferation , Cytokines , Fibroblasts , Humans , Reactive Oxygen Species
7.
Sci Rep ; 8(1): 1540, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367725

ABSTRACT

Visceral fat deposition is associated with impairment of glucose and lipid metabolism while leptin levels are frequently related to subcutaneous fat area. At present, there is considerable controversy regarding the role of visceral adipose tissue accumulation in the development of metabolic syndrome (MS). Here we show the effects of omentectomy on the liver and MS in a diet induced obesity rat model. Our results reveal that undergoing omentectomy previously the establishment of the diet-induced-obesity reduced significantly body weight gain and avoid the development of MS, including non-alcoholic fatty liver disease. Intriguingly, the significantly lower body weight gain was due to decreased food intake. Omentum drives obesity progression through leptin resistance mediated by C-reactive protein, Interleucin (IL)-6 and high lipolysis activity. Omentum removal reversed immediately the increased plasma levels of CRP and IL-6 and gradually food intake, weight gain, and features of MS in diet-induced-obesity. Omentectomy caused no changes in normal-weigh-rats. This report displays causal mechanism by which omentum promotes obesity and propose omentectomy as a promising procedure in MS prevention.


Subject(s)
Appetite , Body Weight , Metabolic Syndrome/prevention & control , Obesity/complications , Obesity/surgery , Omentum/surgery , Surgical Procedures, Operative/methods , Adipogenesis , Animals , C-Reactive Protein/metabolism , Disease Models, Animal , Interleukin-6/metabolism , Leptin/metabolism , Rats , Treatment Outcome
8.
Med Oncol ; 34(7): 122, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560682

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) is a biological process in which a non-motile epithelial cell changes to a mesenchymal state with invasive capacities. However, the EMT program is involved in both physiological and pathological processes. Cancer-associated EMT is known to contribute to increase invasiveness and metastasis, resistance to therapies, and generation of cell populations with stem cell-like characteristics and therefore is deeply involved in tumor progression. This process is finely orchestrated by multiple signaling pathways and regulatory transcriptional networks. The hallmark of EMT is the loss of epithelial surface markers, mainly E-cadherin, and the acquisition of mesenchymal phenotype. These events can be mediated by EMT transcription factors which can cooperate with several enzymes to repress the E-cadherin expression and regulate EMT at the epigenetic and post-translational level. A growing body of evidence indicates that cancer cells can reside in various phenotypic states along the EMT spectrum, where cells can jointly retain epithelial traits with mesenchymal ones. This type of phenotypic plasticity endows cancer cells with tumor-initiating potential. The identification of the signaling pathways and modulators that lead to activation of EMT programs during these disease processes is providing new insights into the plasticity of cellular phenotypes and possible therapeutic interventions.


Subject(s)
Cell Transformation, Neoplastic/pathology , Epithelial-Mesenchymal Transition , Antigens, CD , Cadherins , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Neoplastic Stem Cells
9.
Sci Rep ; 6: 23664, 2016 05 13.
Article in English | MEDLINE | ID: mdl-27173483

ABSTRACT

The aim of this study was to evaluate the role of NADPH oxidase (NADPHox) in the pathogenesis of oxidative phosphorylation (OXPHOS) dysfunction as found in mice fed a high-fat diet (HFD). C57BL/6J mice were distributed in four groups: WT/SCD: six wild-type (WT) mice fed a standard chow diet (SCD); WT/HFD, six WT mice fed a HFD; NOX2(-/-)/SCD, six NADPHox-deficient mice on a SCD; (4) NOX2(-/-)/HFD, six NADPHox-deficient mice on a HFD. After 32 weeks, we studied the liver for: histology; OXPHOS complex activity; fully assembled OXPHOS complexes and their subunits; gene expression of OXPHOS subunits; oxidative and nitrosative stress; and oxidative DNA damage. In the liver of WT/HFD mice, we found a significant decreased in the activity of all OXPHOS complexes, in fully assembled complexes, in the amount of OXPHOS subunits, and in gene expression of mitochondrial DNA-encoded subunits. 8-hydroxy-2'-deoxyguanosine was only increased in mitochondrial DNA. The liver of NOX(-/-)/HFD mice showed mild steatosis but no non-alcoholic steatohepatitis (NASH) lesions were found. OXPHOS activity, OXPHOS subunits, and assembly of subunits into OXPHOS complexes were normal in these mice. We conclude that this study shows that NADPH deficiency protects mice from developing OXPHOS dysfunction and NASH caused by a HFD.


Subject(s)
Diet, High-Fat , NADPH Oxidase 2/metabolism , Oxidative Phosphorylation , 8-Hydroxy-2'-Deoxyguanosine , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , DNA Damage , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Gene Expression , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , NADPH Oxidase 2/deficiency , NADPH Oxidase 2/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , ERRalpha Estrogen-Related Receptor
10.
Dis Model Mech ; 8(2): 183-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25540128

ABSTRACT

Activity of the oxidative phosphorylation system (OXPHOS) is decreased in humans and mice with nonalcoholic steatohepatitis. Nitro-oxidative stress seems to be involved in its pathogenesis. The aim of this study was to determine whether fatty acids are implicated in the pathogenesis of this mitochondrial defect. In HepG2 cells, we analyzed the effect of saturated (palmitic and stearic acids) and monounsaturated (oleic acid) fatty acids on: OXPHOS activity; levels of protein expression of OXPHOS complexes and their subunits; gene expression and half-life of OXPHOS complexes; nitro-oxidative stress; and NADPH oxidase gene expression and activity. We also studied the effects of inhibiting or silencing NADPH oxidase on the palmitic-acid-induced nitro-oxidative stress and subsequent OXPHOS inhibition. Exposure of cultured HepG2 cells to saturated fatty acids resulted in a significant decrease in the OXPHOS activity. This effect was prevented in the presence of a mimic of manganese superoxide dismutase. Palmitic acid reduced the amount of both fully-assembled OXPHOS complexes and of complex subunits. This reduction was due mainly to an accelerated degradation of these subunits, which was associated with a 3-tyrosine nitration of mitochondrial proteins. Pretreatment of cells with uric acid, an antiperoxynitrite agent, prevented protein degradation induced by palmitic acid. A reduced gene expression also contributed to decrease mitochondrial DNA (mtDNA)-encoded subunits. Saturated fatty acids induced oxidative stress and caused mtDNA oxidative damage. This effect was prevented by inhibiting NADPH oxidase. These acids activated NADPH oxidase gene expression and increased NADPH oxidase activity. Silencing this oxidase abrogated totally the inhibitory effect of palmitic acid on OXPHOS complex activity. We conclude that saturated fatty acids caused nitro-oxidative stress, reduced OXPHOS complex half-life and activity, and decreased gene expression of mtDNA-encoded subunits. These effects were mediated by activation of NADPH oxidase. That is, these acids reproduced mitochondrial dysfunction found in humans and animals with nonalcoholic steatohepatitis.


Subject(s)
Fatty Acids/pharmacology , Mitochondria/metabolism , Mitochondria/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Adenosine Triphosphate/metabolism , DNA, Mitochondrial/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Hep G2 Cells , Humans , Mitochondria/drug effects , NADPH Oxidases/metabolism , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Palmitic Acid/pharmacology , Protein Subunits/genetics , Protein Subunits/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
11.
Dis Model Mech ; 7(11): 1287-96, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25261569

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most frequent histological finding in individuals with abnormal liver-function tests in the Western countries. In previous studies, we have shown that oxidative phosphorylation (OXPHOS) is decreased in individuals with NAFLD, but the cause of this mitochondrial dysfunction remains uncertain. The aims of this study were to determine whether feeding mice a high-fat diet (HFD) induces any change in the activity of OXPHOS, and to investigate the mechanisms involved in the pathogenesis of this defect. To that end, 30 mice were distributed between five groups: control mice fed a standard diet, and mice on a HFD and treated with saline solution, melatonin (an antioxidant), MnTBAP (a superoxide dismutase analog) or uric acid (a scavenger of peroxynitrite) for 28 weeks intraperitoneously. In the liver of these mice, we studied histology, activity and assembly of OXPHOS complexes, levels of subunits of these complexes, gene expression of these subunits, oxidative and nitrosative stress, and oxidative DNA damage. In HFD-fed mice, we found nonalcoholic steatohepatitis, increased gene expression of TNFα, IFNγ, MCP-1, caspase-3, TGFß1 and collagen α1(I), and increased levels of 3-tyrosine nitrated proteins. The activity and assembly of all OXPHOS complexes was decreased to about 50-60%. The amount of all studied OXPHOS subunits was markedly decreased, particularly the mitochondrial-DNA-encoded subunits. Gene expression of mitochondrial-DNA-encoded subunits was decreased to about 60% of control. There was oxidative damage to mitochondrial DNA but not to genomic DNA. Treatment of HFD-fed mice with melatonin, MnTBAP or uric acid prevented all changes observed in untreated HFD-fed mice. We conclude that a HFD decreased OXPHOS enzymatic activity owing to a decreased amount of fully assembled complexes caused by a reduced synthesis of their subunits. Antioxidants and antiperoxynitrites prevented all of these changes, suggesting that nitro-oxidative stress played a key role in the pathogenesis of these alterations. Treatment with these agents might prevent the development of NAFLD in humans.


Subject(s)
Diet, High-Fat , Non-alcoholic Fatty Liver Disease/etiology , Oxidative Phosphorylation , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Mice , NADPH Oxidases/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Transcription, Genetic
12.
BMC Biol ; 11: 88, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23915000

ABSTRACT

BACKGROUND: Thiazolidinediones are antidiabetic agents that increase insulin sensitivity but reduce glucose oxidation, state 3 respiration, and activity of complex I of the mitochondrial respiratory chain (MRC). The mechanisms of the latter effects are unclear. The aim of this study was to determine the mechanisms by which pioglitazone (PGZ), a member of the thiazolidinedione class of antidiabetic agents, decreases the activity of the MRC. In isolated mitochondria from mouse liver, we measured the effects of PGZ treatment on MRC complex activities, fully-assembled complex I and its subunits, gene expression of complex I and III subunits, and [3H]PGZ binding to mitochondrial complexes. RESULTS: In vitro, PGZ decreased activity of complexes I and III of the MRC, but in vivo only complex I activity was decreased in mice treated for 12 weeks with 10 mg/kg/day of PGZ. In vitro treatment of isolated liver mitochondria with PGZ disassembled complex I, resulting in the formation of several subcomplexes. In mice treated with PGZ, fully assembled complex I was increased and two additional subcomplexes were found. Formation of supercomplexes CI+CIII2+CIVn and CI+CIII2 decreased in mouse liver mitochondria exposed to PGZ, while formation of these supercomplexes was increased in mice treated with PGZ. Two-dimensional analysis of complex I using blue native/sodium dodecyl sulfate polyacrylamide gel electrophoresis (BN/SDS-PAGE) showed that in vitro PGZ induced the formation of four subcomplexes of 600 (B), 400 (C), 350 (D), and 250 (E) kDa, respectively. Subcomplexes B and C had NADH:dehydrogenase activity, while subcomplexes C and D contained subunits of complex I membrane arm. Autoradiography and coimmunoprecipitation assays showed [3H]PGZ binding to subunits NDUFA9, NDUFB6, and NDUFA6. Treatment with PGZ increased mitochondrial gene transcription in mice liver and HepG2 cells. In these cells, PGZ decreased intracellular ATP content and enhanced gene expression of specific protein 1 and peroxisome-proliferator activated receptor (PPAR)γ coactivator 1α (PGC-1α). CONCLUSIONS: PGZ binds complex I subunits, which induces disassembly of this complex, reduces its activity, depletes cellular ATP, and, in mice and HepG2 cells, upregulates nuclear DNA-encoded gene expression of complex I and III subunits.


Subject(s)
Electron Transport Complex I/metabolism , Mitochondria, Liver/enzymology , Thiazolidinediones/pharmacology , Adenosine Triphosphate/metabolism , Animals , Electron Transport/drug effects , Electron Transport Complex III/metabolism , Enzyme Activation/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , Mice , Mice, Inbred C57BL , Mitochondria, Liver/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Molecular Chaperones/metabolism , Molecular Weight , NADH Dehydrogenase/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pioglitazone , Prohibitins , Protein Subunits/metabolism , Repressor Proteins/metabolism , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Tritium/metabolism , Up-Regulation/drug effects
13.
Endocrinology ; 153(12): 5845-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23093703

ABSTRACT

Mechanisms by which leptin stimulates collagen α(1)(I) [Col1a(I)] gene expression are unclear. The purposes of this study were to identify the trans-acting factors and cis-acting elements in Col1a(I) promoter involved in this effect as well as the pathways that are implicated. In primary cultures of rat hepatic stellate cells (HSCs), we measured the effects of leptin on Col1a(I) gene and protein expression and on the binding of nuclear proteins to the Col1a(I) promoter. We found that leptin increased Col1a(I) gene and protein expression in activated HSCs. Transient transfections showed that leptin exerted its effects through elements located between -220 and -112 bp of the Col1a(I) promoter. Gel retardation assays demonstrated that leptin induced the binding of transcription factors specific protein (Sp)-1 and Sp3 to two elements located between -161 and -110 bp of the Col1a(I) promoter. Leptin-induced Sp1/Sp3 phosphorylation, but this effect was suppressed by inhibiting or silencing Janus kinase-2, phosphatidylinositol-3-kinase, nonphagocytic adenine dinucleotide phosphate (NADPH) oxidase, or ERK1/2, by the use of antioxidants or catalase, or by preventing protein-aldehyde adduct formation. Leptin provoked oxidative stress, aldehyde-protein adduct formation, and increased gene expression of some components of the NADPH oxidase complex. In conclusion, in HSCs, leptin up-regulates Col1a(I) gene expression after activating NADPH oxidase, inducing oxidative stress, aldehyde-protein adduct formation, and ERK1/2 phosphorylation, which in turn activates Sp1/Sp3 and provokes the binding of these two factors to regulatory elements located between -161 and -110 bp of the Col1a(I) promoter. These findings may contribute to a better understanding of mechanisms involved in the leptin-induced liver fibrosis.


Subject(s)
Collagen Type I/metabolism , Gene Expression Regulation , Hepatic Stellate Cells/cytology , Leptin/metabolism , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Animals , Cells, Cultured , Fibrosis/pathology , Glutathione/metabolism , Male , Models, Genetic , Oxidative Stress , Phosphorylation , Promoter Regions, Genetic , RNA Interference , Rats , Rats, Sprague-Dawley
14.
J Biol Chem ; 287(23): 19564-73, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22493491

ABSTRACT

Insulin resistance is a risk factor for non-response to interferon/ribavirin therapy in patients with chronic hepatitis C. The aim of this study was to determine the role played by protein-tyrosine phosphatases (PTPs) in the absence of interferon-α (IFNα) response associated with insulin resistance. We induced insulin resistance by silencing IRS-2 or by treating HepG2 cells with tumor necrosis factor-α (TNFα) and analyzed insulin response by evaluating Akt phosphorylation and IFNα response by measuring Stat-1 tyrosine phosphorylation and 2',5'-oligoadenylate synthase and myxovirus resistance gene expression. The response to IFNα was also measured in insulin-resistant obese mice (high fat diet and ob/ob mice) untreated and treated with metformin. Silencing IRS-2 mRNA induces insulin resistance and inhibits IFNα response. Likewise, TNFα suppresses insulin and IFNα response. Treatment of cells with pervanadate and knocking down PTP-1B restores insulin and IFNα response. Both silencing IRS-2 and TNFα treatment increase PTP and PTP-1B activity. Metformin inhibits PTP and improves IFNα response in insulin-resistant cells. Insulin-resistant ob/ob mice have increased PTP-1B gene expression and activity in the liver and do not respond to IFNα administration. Treatment with metformin improves this response. In HepG2 cells, insulin resistance provokes IFNα resistance, which is associated with an increased PTP-1B activity in the liver. Inhibition of PTP-1B activity with pervanadate and metformin or knocking down PTP-1B reestablishes IFNα response. Likewise, metformin decreases PTP-1B activity and improves response to IFNα in insulin-resistant obese mice. The use of PTP-1B inhibitors may improve the response to IFNα/ribavirin therapy.


Subject(s)
Antiviral Agents/pharmacology , Insulin Resistance , Interferon-alpha/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Gene Silencing , Hep G2 Cells , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/enzymology , Hepatitis C, Chronic/genetics , Humans , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Metformin/pharmacology , Mice , Mice, Obese , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Proto-Oncogene Proteins c-akt , Ribavirin/pharmacology , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vanadates/pharmacology
16.
J Pineal Res ; 51(1): 113-23, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21355880

ABSTRACT

In previous studies, we have shown that mitochondrial respiratory chain (MRC) activity is decreased in patients with nonalcoholic steatohepatitis and in ob/ob mice and that peroxynitrite plays a pathogenic role. The present study examined whether melatonin, a peroxynitrite scavenger, prevents: (i) the in vitro effects of peroxynitrite on normal mitochondrial proteins and (ii) the development of nonalcoholic liver disease, MRC dysfunction and proteomic changes found in the mitochondrial complexes from ob/ob mice. We studied MRC activity, assembly of mitochondrial complexes and its subunits in normal mitochondrial proteins exposed to peroxynitrite in the absence and presence of melatonin. The same studies were done in mitochondrial proteins from ob/ob mice untreated and treated with melatonin. Preincubation of mitochondrial proteins from wild-type mice with melatonin prevented 3-tyrosine nitration of these proteins, eliminated the reduction in the MRC activity, the defect in the assembly of mitochondrial complexes and degradation of their subunits induced by peroxynitrite in vitro. Moreover, treatment of ob/ob mice with 10 mg/kg/day melatonin for 12 wk reduced oxidative and nitrosative stress, prevented the loss of MRC activity, protected their complexes and subunits from degradation, and favored assembling of mitochondrial complexes. In addition, this treatment improved fatty liver, decreased hepatic triglyceride concentration and increased apolipoprotein B100 in liver tissue. In conclusion, melatonin prevents the effects of peroxynitrite on mitochondrial proteins in vitro and administration of melatonin to ob/ob mice normalizes liver morphology, mitochondrial dysfunction and assembly of MRC complexes.


Subject(s)
Electron Transport Chain Complex Proteins/metabolism , Liver/drug effects , Melatonin/pharmacology , Animals , Blotting, Western , Body Weight/drug effects , Fatty Liver/metabolism , Fatty Liver/prevention & control , Histocytochemistry , Liver/cytology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Nitrates/metabolism , Oxidative Stress/drug effects , Peroxynitrous Acid/pharmacology , Protein Subunits , Tyrosine/metabolism
17.
J Proteome Res ; 9(5): 2450-9, 2010 May 07.
Article in English | MEDLINE | ID: mdl-20222668

ABSTRACT

We investigate the cause of the low activity of mitochondrial complex I found in ob/ob mice with nonalcoholic fatty liver disease. In mitochondrial proteins from ob/ob mice, we assessed complex I activity, fully assembled complex I, and its subunits, oxygen consumption, gene expression of complex I subunits, and oxidative damage to DNA. In mitochondrial proteins from the liver of ob/ob mice, complex I activity, fully assembly of this complex and complex I subunits were markedly reduced. Likewise, gene expression of mitochondrial DNA-encoded subunits was significantly decreased in obese mice, but not nuclear DNA-encoded subunits. Treatment of obese mice with uric acid, anti-TNFalpha antibody or a mimic of manganese superoxide dismutase normalized all these abnormalities. "In vitro" addition of peroxynitrite to mitochondrial proteins from wild-type mice reproduced the abnormalities found in ob/ob mice (decreased complex I activity, the amount of fully assembled complex I, and its subunits, and mitochondrial oxygen consumption). Low activity of complex I found in ob/ob mice can be ascribed to a reduced amount of fully assembled complex, which may be attributed to degradation and reduced synthesis of its subunits by peroxynitrite. Exposure of mitochondrial proteins from normal mice to peroxynitrite reproduced the proteomic abnormalities present in ob/ob mice.


Subject(s)
Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Fatty Liver/metabolism , Peroxynitrous Acid/pharmacology , Animals , DNA Damage , DNA, Mitochondrial/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/genetics , Fatty Liver/pathology , Leptin/genetics , Liver/chemistry , Liver/drug effects , Male , Metalloporphyrins , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Mitochondrial Membranes/metabolism , Oxidative Stress , Prohibitins , Repressor Proteins/metabolism
18.
Cell Physiol Biochem ; 24(3-4): 271-82, 2009.
Article in English | MEDLINE | ID: mdl-19710542

ABSTRACT

UNLABELLED: The aims of this study were to determine whether fibronectin increases survival of hepatic stellate cells (HSCs) in starving conditions, and to identify the signal transduction pathways involved in this effect. METHODS: Primary culture of rat HSCs were plated on fibronectin-uncoated or coated culture wells, and grown in the presence of 0.2% or 20% fetal calf serum. Cell apoptosis was measured by an ELISA procedure. Signal transduction pathways were analyzed by inhibiting major intracellular transduction pathways with appropriated inhibitors and by detecting phosphorylated proteins. RESULTS: Fibronectin increased survival of serum deprived HSCs. This effect was abrogated by the presence of the RGD peptide, by silencing FAK expression, and by inhibiting PI3K with LY294002 or wortmannin. Growth of HSCs on fibronectin induced integrin alpha5beta1 expression, tyr397, ser473, and ser136 phosphorylation of FAK, Akt, and Bad, respectively, and the binding of phosphorylated Bad to 14-3-3 proteins. Likewise, fibronectin increased Bcl2/Bax ratio and reduced release of mitochondrial cytochrome c into the cytoplasm, formation of apoptosome, and caspase 9 and 3 activity. These effects were avoided by treatment of cells with PI3K inhibitors. CONCLUSION: Fibronectin increases survival of HSCs via a pathway involving integrin alpha5beta1 receptors, FAK, PI3K, Akt and proteins of Bcl2 family.


Subject(s)
Fibronectins/metabolism , Hepatic Stellate Cells/metabolism , 14-3-3 Proteins/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Caspase 9/metabolism , Cell Culture Techniques , Cell Death , Cell Separation/methods , Cell Survival , Cells, Cultured , Chromones/pharmacology , Coated Materials, Biocompatible/metabolism , Culture Media, Serum-Free , Enzyme Activation , Enzyme Inhibitors/pharmacology , Integrin alpha5beta1/metabolism , Male , Morpholines/pharmacology , Oligopeptides/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors , Transfection , bcl-2-Associated X Protein/metabolism
19.
J Hepatol ; 50(1): 128-39, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19014879

ABSTRACT

BACKGROUND/AIMS: To determine the effects of IFNalpha on MMP-13 gene expression in primary culture of hepatic stellate cells. METHODS: We measured MMP-13 mRNA, MMP-13 protein, MMP-13 luciferase activity, binding of AP1 and PEA3 to DNA, and binding of PEA3 to Jak1 and Stat1. RESULTS: IFNalpha increased MMP-13 mRNA, MMP-13 protein, and luciferase activity in cells transfected either with a luciferase plasmid driven by the MMP-13 promoter or with the same plasmid in which the AP1 binding site has been mutated. IFNalpha induced the binding of nuclear proteins to a radiolabeled PEA3 probe, but not to a AP1 probe. Supershift assays demonstrated that PEA3 and Stat1 are implicated in the formation of this complex. Immunoprecipitation assays showed that PEA3 interacts physically with Stat1 and that IFNalpha treatment increases this interaction. Downregulation of PEA3 or JAK1 with appropriated siRNAs or mutation of the PEA3 binding site in the MMP-13 promoter abrogated the effects of IFNalpha on MMP-13 gene expression. Finally, IFNalpha induced the binding of PEA3 to JAK1, as well as PEA3 tyrosine and serine phosphorylation. CONCLUSIONS: IFNalpha determines the binding of PEA3 to JAK1 and its tyrosine phosphorylation. Activated PEA3 binds to MMP-13 promoter and activates its expression.


Subject(s)
Antiviral Agents/pharmacology , Gene Expression/drug effects , Hepatic Stellate Cells/metabolism , Interferon-alpha/pharmacology , Matrix Metalloproteinase 13/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/drug effects , Janus Kinase 1/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor/metabolism , Serine/metabolism , Signal Transduction/drug effects
20.
Hepatology ; 46(2): 414-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17654601

ABSTRACT

UNLABELLED: Insulin resistance is present in almost all patients with nonalcoholic steatohepatitis (NAFLD), and mitochondrial dysfunction likely plays a critical role in the progression of fatty liver into nonalcoholic steatohepatitis. Rosiglitazone, a selective ligand of peroxisome proliferator-activated receptor gamma (PPARgamma), is an insulin sensitizer drug that has been used in a number of insulin-resistant conditions, including NAFLD. The aim of this study was to analyze the effects of rosiglitazone on the liver histology and mitochondrial function in a model of NAFLD. All studies were carried out in wild-type and leptin-deficient (ob/ob) C57BL/6J mice. Ob/ob mice were treated with 1 mg/kg/day, and activity of mitochondrial respiratory chain (MRC), beta-oxidation, lipid peroxidation, glutathione content in mitochondria, and 3-tyrosine-nitrated proteins in mitochondria were measured. In addition, histological and ultrastructural changes induced by rosiglitazone were also noted. Rosiglitazone treatment increased liver steatosis, particularly microvesicular steatosis. In these animals, mitochondria were markedly swollen with cristae peripherally placed. In ob/ob mice, this drug increased PPARgamma protein expression and lipid peroxide content in liver tissue and decreased glutathione concentration in mitochondria. Rosiglitazone suppressed the activity of complex I of the MRC in ob/ob mice, but did not affect beta-oxidation. 3-Tyrosine nitrated mitochondrial proteins, significantly increased in ob/ob mice, were not modified by rosiglitazone treatment. CONCLUSION: Treatment of ob/ob mice with rosiglitazone did not reverse histological lesions of NAFLD or improve MRC activity. On the contrary, rosiglitazone reduced activity of complex I and increased oxidative stress and liver steatosis.


Subject(s)
Liver/drug effects , Mitochondria, Liver/drug effects , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Body Weight/drug effects , Fatty Liver/drug therapy , Fatty Liver/pathology , Lipid Peroxidation/drug effects , Liver/pathology , Liver/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microscopy, Electron , Mitochondria, Liver/metabolism , PPAR gamma/analysis , Rosiglitazone , Tyrosine/analogs & derivatives , Tyrosine/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...