Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 261
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(12)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38928146

ABSTRACT

Mitochondrial quality control is essential in mitochondrial function. To examine the importance of Parkin-dependent mechanisms in mitochondrial quality control, we assessed the impact of modulating Parkin on proteome flux and mitochondrial function in a context of reduced mtDNA fidelity. To accomplish this, we crossed either the Parkin knockout mouse or ParkinW402A knock-in mouse lines to the Polg mitochondrial mutator line to generate homozygous double mutants. In vivo longitudinal isotopic metabolic labeling was followed by isolation of liver mitochondria and synaptic terminals from the brain, which are rich in mitochondria. Mass spectrometry and bioenergetics analysis were assessed. We demonstrate that slower mitochondrial protein turnover is associated with loss of mtDNA fidelity in liver mitochondria but not synaptic terminals, and bioenergetic function in both tissues is impaired. Pathway analysis revealed loss of mtDNA fidelity is associated with disturbances of key metabolic pathways, consistent with its association with metabolic disorders and neurodegeneration. Furthermore, we find that loss of Parkin leads to exacerbation of Polg-driven proteomic consequences, though it may be bioenergetically protective in tissues exhibiting rapid mitochondrial turnover. Finally, we provide evidence that, surprisingly, dis-autoinhibition of Parkin (ParkinW402A) functionally resembles Parkin knockout and fails to rescue deleterious Polg-driven effects. Our study accomplishes three main outcomes: (1) it supports recent studies suggesting that Parkin dependence is low in response to an increased mtDNA mutational load, (2) it provides evidence of a potential protective role of Parkin insufficiency, and (3) it draws into question the therapeutic attractiveness of enhancing Parkin function.


Subject(s)
DNA Polymerase gamma , DNA, Mitochondrial , Mice, Knockout , Mutation , Ubiquitin-Protein Ligases , Animals , DNA Polymerase gamma/genetics , DNA Polymerase gamma/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Mice , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Proteomics/methods , Proteome/metabolism , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics
2.
FASEB J ; 36(1): e22091, 2022 01.
Article in English | MEDLINE | ID: mdl-34919756

ABSTRACT

Hepatoencephalopathy due to combined oxidative phosphorylation deficiency type 1 (COXPD1) is a recessive mitochondrial translation disorder caused by mutations in GFM1, a nuclear gene encoding mitochondrial elongation factor G1 (EFG1). Patients with COXPD1 typically present hepatoencephalopathy early after birth with rapid disease progression, and usually die within the first few weeks or years of life. We have generated two different mouse models: a Gfm1 knock-in (KI) harboring the p.R671C missense mutation, found in at least 10 patients who survived more than 1 year, and a Gfm1 knock-out (KO) model. Homozygous KO mice (Gfm1-/- ) were embryonically lethal, whereas homozygous KI (Gfm1R671C/R671C ) mice were viable and showed normal growth. R671C mutation in Gfm1 caused drastic reductions in the mitochondrial EFG1 protein content in different organs. Six- to eight-week-old Gfm1R671C/R671C mice showed partial reductions of in organello mitochondrial translation and respiratory complex IV enzyme activity in the liver. Compound heterozygous Gfm1R671C/- showed a more pronounced decrease of EFG1 protein in liver and brain mitochondria, as compared with Gfm1R671C/R671C mice. At 8 weeks of age, their mitochondrial translation rates were significantly reduced in both tissues. Additionally, Gfm1R671C/- mice showed combined oxidative phosphorylation deficiency (reduced complex I and IV enzyme activities in liver and brain), and blue native polyacrylamide gel electrophoresis analysis revealed lower amounts of both affected complexes. We conclude that the compound heterozygous Gfm1R671C/- mouse presents a clear dysfunctional molecular phenotype, showing impaired mitochondrial translation and combined respiratory chain dysfunction, making it a suitable animal model for the study of COXPD1.


Subject(s)
Hepatic Encephalopathy/metabolism , Metabolism, Inborn Errors/metabolism , Mitochondria, Liver/metabolism , Mitochondrial Proteins/metabolism , Mutation, Missense , Oxidative Phosphorylation , Peptide Elongation Factor G/metabolism , Protein Biosynthesis , Amino Acid Substitution , Animals , Disease Models, Animal , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Hepatic Encephalopathy/genetics , Metabolism, Inborn Errors/genetics , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondrial Proteins/genetics , Peptide Elongation Factor G/genetics
3.
Mol Cell ; 57(1): 69-82, 2015 Jan 08.
Article in English | MEDLINE | ID: mdl-25482509

ABSTRACT

Proapoptotic BCL-2 proteins converge upon the outer mitochondrial membrane (OMM) to promote mitochondrial outer membrane permeabilization (MOMP) and apoptosis. Here we investigated the mechanistic relationship between mitochondrial shape and MOMP and provide evidence that BAX requires a distinct mitochondrial size to induce MOMP. We utilized the terminal unfolded protein response pathway to systematically define proapoptotic BCL-2 protein composition after stress and then directly interrogated their requirement for a productive mitochondrial size. Complementary biochemical, cellular, in vivo, and ex vivo studies reveal that Mfn1, a GTPase involved in mitochondrial fusion, establishes a mitochondrial size that is permissive for proapoptotic BCL-2 family function. Cells with hyperfragmented mitochondria, along with size-restricted OMM model systems, fail to support BAX-dependent membrane association and permeabilization due to an inability to stabilize BAXα9·membrane interactions. This work identifies a mechanistic contribution of mitochondrial size in dictating BAX activation, MOMP, and apoptosis.


Subject(s)
GTP Phosphohydrolases/genetics , Mitochondria, Liver/genetics , Mitochondrial Membranes/metabolism , Organelle Shape/genetics , bcl-2-Associated X Protein/genetics , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , GTP Phosphohydrolases/metabolism , Gene Expression Regulation , Membrane Potential, Mitochondrial/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria, Liver/metabolism , Mitochondria, Liver/ultrastructure , Mitochondrial Dynamics/genetics , Mitochondrial Membranes/ultrastructure , Permeability , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Unilamellar Liposomes/chemistry , Unilamellar Liposomes/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
4.
J Biol Chem ; 297(6): 101388, 2021 12.
Article in English | MEDLINE | ID: mdl-34762911

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT) converts nicotinamide to NAD+. As low hepatic NAD+ levels have been linked to the development of nonalcoholic fatty liver disease, we hypothesized that ablation of hepatic Nampt would affect susceptibility to liver injury in response to diet-induced metabolic stress. Following 3 weeks on a low-methionine and choline-free 60% high-fat diet, hepatocyte-specific Nampt knockout (HNKO) mice accumulated less triglyceride than WT littermates but had increased histological scores for liver inflammation, necrosis, and fibrosis. Surprisingly, liver injury was also observed in HNKO mice on the purified control diet. This HNKO phenotype was associated with decreased abundance of mitochondrial proteins, especially proteins involved in oxidoreductase activity. High-resolution respirometry revealed lower respiratory capacity in purified control diet-fed HNKO liver. In addition, fibrotic area in HNKO liver sections correlated negatively with hepatic NAD+, and liver injury was prevented by supplementation with NAD+ precursors nicotinamide riboside and nicotinic acid. MS-based proteomic analysis revealed that nicotinamide riboside supplementation rescued hepatic levels of oxidoreductase and OXPHOS proteins. Finally, single-nucleus RNA-Seq showed that transcriptional changes in the HNKO liver mainly occurred in hepatocytes, and changes in the hepatocyte transcriptome were associated with liver necrosis. In conclusion, HNKO livers have reduced respiratory capacity, decreased abundance of mitochondrial proteins, and are susceptible to fibrosis because of low NAD+ levels. Our data suggest a critical threshold level of hepatic NAD+ that determines the predisposition to liver injury and supports that NAD+ precursor supplementation can prevent liver injury and nonalcoholic fatty liver disease progression.


Subject(s)
Hepatocytes/metabolism , Mitochondria, Liver/metabolism , NAD/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Cytokines/deficiency , Cytokines/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/genetics , NAD/genetics , Nicotinamide Phosphoribosyltransferase/deficiency , Nicotinamide Phosphoribosyltransferase/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Phosphorylation , Phenotype
5.
Arterioscler Thromb Vasc Biol ; 41(2): 734-754, 2021 02.
Article in English | MEDLINE | ID: mdl-33297749

ABSTRACT

OBJECTIVE: NFU1 is a mitochondrial iron-sulfur scaffold protein, involved in iron-sulfur assembly and transfer to complex II and LAS (lipoic acid synthase). Patients with the point mutation NFU1G208C and CRISPR/CAS9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9)-generated rats develop mitochondrial dysfunction leading to pulmonary arterial hypertension. However, the mechanistic understanding of pulmonary vascular proliferation due to a single mutation in NFU1 remains unresolved. Approach and Results: Quantitative proteomics of isolated mitochondria showed the entire phenotypic transformation of NFU1G206C rats with a disturbed mitochondrial proteomic landscape, involving significant changes in the expression of 208 mitochondrial proteins. The NFU1 mutation deranged the expression pattern of electron transport proteins, resulting in a significant decrease in mitochondrial respiration. Reduced reliance on mitochondrial respiration amplified glycolysis in pulmonary artery smooth muscle cell (PASMC) and activated GPD (glycerol-3-phosphate dehydrogenase), linking glycolysis to oxidative phosphorylation and lipid metabolism. Decreased PDH (pyruvate dehydrogenase) activity due to the lipoic acid shortage is compensated by increased fatty acid metabolism and oxidation. PASMC became dependent on extracellular fatty acid sources due to upregulated transporters such as CD36 (cluster of differentiation 36) and CPT (carnitine palmitoyltransferase)-1. Finally, the NFU1 mutation produced a dysregulated antioxidant system in the mitochondria, leading to increased reactive oxygen species levels. PASMC from NFU1 rats showed apoptosis resistance, increased anaplerosis, and attained a highly proliferative phenotype. Attenuation of mitochondrial reactive oxygen species by mitochondrial-targeted antioxidant significantly decreased PASMC proliferation. CONCLUSIONS: The alteration in iron-sulfur metabolism completely transforms the proteomic landscape of the mitochondria, leading toward metabolic plasticity and redistribution of energy sources to the acquisition of a proliferative phenotype by the PASMC.


Subject(s)
Apoptosis , Cell Proliferation , Cellular Reprogramming , Energy Metabolism , Mitochondria, Liver/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Point Mutation , Animals , Cells, Cultured , Fatty Acids/metabolism , Female , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Proteome , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction
6.
J Biol Chem ; 295(50): 17310-17322, 2020 12 11.
Article in English | MEDLINE | ID: mdl-33037071

ABSTRACT

In addition to their well-known role in the control of cellular proliferation and cancer, cell cycle regulators are increasingly identified as important metabolic modulators. Several GWAS have identified SNPs near CDKN2A, the locus encoding for p16INK4a (p16), associated with elevated risk for cardiovascular diseases and type-2 diabetes development, two pathologies associated with impaired hepatic lipid metabolism. Although p16 was recently shown to control hepatic glucose homeostasis, it is unknown whether p16 also controls hepatic lipid metabolism. Using a combination of in vivo and in vitro approaches, we found that p16 modulates fasting-induced hepatic fatty acid oxidation (FAO) and lipid droplet accumulation. In primary hepatocytes, p16-deficiency was associated with elevated expression of genes involved in fatty acid catabolism. These transcriptional changes led to increased FAO and were associated with enhanced activation of PPARα through a mechanism requiring the catalytic AMPKα2 subunit and SIRT1, two known activators of PPARα. By contrast, p16 overexpression was associated with triglyceride accumulation and increased lipid droplet numbers in vitro, and decreased ketogenesis and hepatic mitochondrial activity in vivo Finally, gene expression analysis of liver samples from obese patients revealed a negative correlation between CDKN2A expression and PPARA and its target genes. Our findings demonstrate that p16 represses hepatic lipid catabolism during fasting and may thus participate in the preservation of metabolic flexibility.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Fatty Acids/metabolism , Liver/metabolism , Mitochondria, Liver/metabolism , PPAR alpha/metabolism , Signal Transduction , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Fatty Acids/genetics , Genome-Wide Association Study , Humans , Lipid Droplets/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Obesity/genetics , Obesity/metabolism , Oxidation-Reduction , PPAR alpha/genetics , Sirtuin 1/genetics
7.
Int J Mol Sci ; 22(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498298

ABSTRACT

Several genetic variants in the mitochondrial genome (mtDNA), including ancient polymorphisms, are associated with chronic inflammatory conditions, but investigating the functional consequences of such mtDNA polymorphisms in humans is challenging due to the influence of many other polymorphisms in both mtDNA and the nuclear genome (nDNA). Here, using the conplastic mouse strain B6-mtFVB, we show that in mice, a maternally inherited natural mutation (m.7778G > T) in the mitochondrially encoded gene ATP synthase 8 (mt-Atp8) of complex V impacts on the cellular metabolic profile and effector functions of CD4+ T cells and induces mild changes in oxidative phosphorylation (OXPHOS) complex activities. These changes culminated in significantly lower disease susceptibility in two models of inflammatory skin disease. Our findings provide experimental evidence that a natural variation in mtDNA influences chronic inflammatory conditions through alterations in cellular metabolism and the systemic metabolic profile without causing major dysfunction in the OXPHOS system.


Subject(s)
DNA, Mitochondrial/genetics , Epidermolysis Bullosa Acquisita/genetics , Lymphocytes/metabolism , Polymorphism, Single Nucleotide , Animals , Cells, Cultured , Cytokines/metabolism , Epidermolysis Bullosa Acquisita/metabolism , Mice , Mice, Inbred C57BL , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Mitochondrial Proton-Translocating ATPases/genetics
8.
J Biol Chem ; 294(19): 7810-7820, 2019 05 10.
Article in English | MEDLINE | ID: mdl-30923124

ABSTRACT

Aerobic glycolysis and mitochondrial dysfunction are key metabolic features of cancer cells, but their interplay during cancer development remains unclear. We previously reported that human hepatoma cells with mitochondrial defects exhibit down-regulated lactate dehydrogenase subunit B (LDHB) expression. Here, using several molecular and biochemical assays and informatics analyses, we investigated how LDHB suppression regulates mitochondrial respiratory activity and contributes to liver cancer progression. We found that transcriptional LDHB down-regulation is an upstream event during suppressed oxidative phosphorylation. We also observed that LDHB knockdown increases inhibitory phosphorylation of pyruvate dehydrogenase (PDH) via lactate-mediated PDH kinase (PDK) activation and thereby attenuates oxidative phosphorylation activity. Interestingly, monocarboxylate transporter 1 was the major lactate transporter in hepatoma cells, and its expression was essential for PDH phosphorylation by modulating intracellular lactate levels. Finally, bioinformatics analysis of the hepatocellular carcinoma cohort from The Cancer Genome Atlas revealed that a low LDHB/LDHA ratio is statistically significantly associated with poor prognostic outcomes. A low ratio was also associated with a significant enrichment in glycolysis genes and negatively correlated with PDK1 and 2 expression, supporting a close link between LDHB suppression and the PDK-PDH axis. These results suggest that LDHB suppression is a key mechanism that enhances glycolysis and is critically involved in the maintenance and propagation of mitochondrial dysfunction via lactate release in liver cancer progression.


Subject(s)
Acidosis, Lactic/enzymology , Carcinoma, Hepatocellular/enzymology , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lactate Dehydrogenases/biosynthesis , Liver Neoplasms/enzymology , Mitochondria, Liver/enzymology , Neoplasm Proteins/blood , Oxidative Phosphorylation , Acidosis, Lactic/genetics , Acidosis, Lactic/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Lactate Dehydrogenases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Neoplasm Proteins/genetics
9.
J Biol Chem ; 294(33): 12359-12369, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31235522

ABSTRACT

Excessive alcohol consumption induces intestinal dysbiosis of the gut microbiome and reduces gut epithelial integrity. This often leads to portal circulation-mediated translocation of gut-derived microbial products, such as lipopolysaccharide (LPS), to the liver, where these products engage Toll-like receptor 4 (TLR4) and initiate hepatic inflammation, which promotes alcoholic liver disease (ALD). Although the key self-destructive process of autophagy has been well-studied in hepatocytes, its role in macrophages during ALD pathogenesis remains elusive. Using WT and myeloid cell-specific autophagy-related 7 (Atg7) knockout (Atg7ΔMye) mice, we found that chronic ethanol feeding for 6 weeks plus LPS injection enhances serum alanine aminotransferase and IL-1ß levels and augments hepatic C-C motif chemokine ligand 5 (CCL5) and C-X-C motif chemokine ligand 10 (CXCL10) expression in WT mice, a phenotype that was further exacerbated in Atg7ΔMye mice. Atg7ΔMye macrophages exhibited defective mitochondrial respiration and displayed elevated mitochondrial reactive oxygen species production and inflammasome activation relative to WT cells. Interestingly, compared with WT cells, Atg7ΔMye macrophages also had a drastically increased abundance and nuclear translocation of interferon regulatory factor 1 (IRF1) after LPS stimulation. Mechanistically, LPS induced co-localization of IRF1 with the autophagy adaptor p62 and the autophagosome, resulting in subsequent IRF1 degradation. However, upon p62 silencing or Atg7 deletion, IRF1 started to accumulate in autophagy-deficient macrophages and translocated into the nucleus, where it induced CCL5 and CXCL10 expression. In conclusion, macrophage autophagy protects against ALD by promoting IRF1 degradation and removal of damaged mitochondria, limiting macrophage activation and inflammation.


Subject(s)
Autophagic Cell Death , Chemical and Drug Induced Liver Injury/metabolism , Ethanol/adverse effects , Interferon Regulatory Factor-1/metabolism , Macrophages/metabolism , Mitochondria, Liver/metabolism , Proteolysis , Animals , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Ethanol/pharmacology , Interferon Regulatory Factor-1/genetics , Lipopolysaccharides/toxicity , Liver/metabolism , Liver/pathology , Macrophages/pathology , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology
10.
Cell Physiol Biochem ; 54(4): 517-537, 2020 May 20.
Article in English | MEDLINE | ID: mdl-32428391

ABSTRACT

BACKGROUND/AIMS: Src kinase family members, including c-Src, are involved in numerous signaling pathways and have been observed inside different cellular compartments. Notably, c-Src modulates carbohydrate and fatty acid metabolism and is involved in the metabolic rewiring of cancer cells. This kinase is found within mitochondria where it targets different proteins to impact on the organelle functions and overall metabolism. Surprisingly, no global metabolic characterization of Src has been performed although c-Src knock-out mice have been available for 30 years. Considering that c-Src is sensitive to various metabolites, c-Src might represent a crucial player in metabolic adjustments induced by nutrient stress. The aim of this work was to characterize the impact of c-Src on mitochondrial activity and overall metabolism using multi-omic characterization. METHODS: Src+/+ and Src-/- mice were fed ad libitum or fasted during 24h and were then analyzed using multi-omics. RESULTS: We observed that deletion of c-Src is linked to lower phosphorylation of Y412-NDUFA8, inhibition of oxygen consumption and accumulation of metabolites involved in glycolysis, TCA cycle and amino acid metabolism in mice fed ad libitum. Finally, metabolomics and (phosphotyrosine) proteomics are differently impacted by Src according to nutrient availability. CONCLUSION: The findings presented here highlight that c-Src reduces mitochondrial metabolism and impacts the metabolic adjustment induced by nutrient stress.


Subject(s)
Mitochondria/metabolism , Phosphotyrosine/metabolism , Proteome/metabolism , src-Family Kinases/metabolism , Animals , Brain/metabolism , Chromatography, Liquid , Citric Acid Cycle/genetics , Gas Chromatography-Mass Spectrometry , Glycolysis/genetics , Kidney/metabolism , Mice , Mice, Knockout , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Nutrients/metabolism , Phosphorylation , Phosphotyrosine/genetics , Proteomics , Tandem Mass Spectrometry , src-Family Kinases/genetics
11.
Am J Pathol ; 189(3): 580-589, 2019 03.
Article in English | MEDLINE | ID: mdl-30553835

ABSTRACT

Despite the growing global burden of alcoholic liver diseases, therapeutic options are limited, and novel targets are urgently needed. Accumulating evidence suggests that mitochondria adapt in response to ethanol and formation of megamitochondria in the livers of patients is recognized as a hallmark of alcoholic liver diseases. The processes involved in ethanol-induced hepatic mitochondrial changes, the impact on mitochondria-shaping proteins, and the significance of megamitochondria formation remain unknown. In this study, we investigated the mitochondrial and cellular response to alcohol in hepatoma cell line VL-17A. The mitochondrial architecture rapidly changed after 3 or 14 days of ethanol exposure with double-pronged presentation of hyperfragmentation and megamitochondria, and cell growth was inhibited. Dynamin-1-like protein (Drp1) was identified as the main mediator driving these mitochondrial alterations, and its genetic inactivation was determined to foster megamitochondria development, preserving the capacity of the cells to grow despite alcohol toxicity. The role of Drp1 in mediating megamitochondria formation in mice with liver-specific inactivation of Drp1 was further confirmed. Finally, when these mice were fed with ethanol, the presentation of hepatic megamitochondria was exacerbated compared with wild type fed with the same diet. Ethanol-induced toxicity was also reduced. Our study demonstrates that megamitochondria formation is mediated by Drp1, and this phenomenon is a beneficial adaptive response during alcohol-induced hepatotoxicity.


Subject(s)
Dynamins/metabolism , Liver Diseases, Alcoholic/metabolism , Mitochondria, Liver/metabolism , Animals , Cell Line, Tumor , Dynamins/genetics , Ethanol/adverse effects , Ethanol/pharmacology , Humans , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/pathology , Mice , Mice, Transgenic , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology
12.
Am J Pathol ; 189(3): 552-567, 2019 03.
Article in English | MEDLINE | ID: mdl-30553838

ABSTRACT

Alcoholic liver disease is associated with high morbidity and mortality, and treatment options are limited to date. Augmenter of liver regeneration (ALR) may protect against hepatic injury from chemical poisons, including ethanol. Autophagy appears to positively influence survival in cases of liver dysfunction, although the mechanisms are poorly understood. Herein, we investigated effects of ALR-induced autophagy in vitro and in vivo in an ethanol-induced model of acute liver injury. Decreased serum levels of alanine aminotransferase and aspartate aminotransferase and reduced histologic lesions revealed that mice overexpressing ALR experienced less liver damage than wild-type. ALR-knockdown mice experienced more severe liver damage than wild-type. ALR-transfected HepG2 cells showed increased survival rates, improved maintenance of mitochondrial membrane potential, and increased ATP levels after ethanol treatment. The observed protection was associated with up-regulation of autophagy-markers, including light chain 3II, beclin-1, and autophagy-related gene 5, and down-regulation of p62 by ALR. Autophagy was inhibited in ALR-knockdown mice and HepG2 cells, and autophagy inhibitor bafilomycin A1 attenuated the protective effects of ALR. Results showed phosphorylated mammalian target of rapamycin (mTOR) was down-regulated when ALR was overexpressed and up-regulated when ALR was knocked down. These data show that ALR is protective against ethanol-induced acute liver injury by promoting autophagy, probably via repressing the mTOR pathway. These results have potential implications for the clinical treatment of alcoholic liver disease patients.


Subject(s)
Acute Lung Injury/prevention & control , Autophagy/drug effects , Ethanol/adverse effects , Liver Diseases, Alcoholic/prevention & control , Liver Regeneration/drug effects , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Ethanol/pharmacology , Hep G2 Cells , Humans , Liver Diseases, Alcoholic/genetics , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Regeneration/genetics , Macrolides/pharmacology , Male , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/genetics , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
13.
Arch Biochem Biophys ; 692: 108535, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32781052

ABSTRACT

NAD(P)+ transhydrogenase (NNT) is located in the inner mitochondrial membrane and catalyzes a reversible hydride transfer between NAD(H) and NADP(H) that is coupled to proton translocation between the intermembrane space and mitochondrial matrix. NNT activity has an essential role in maintaining the NADPH supply for antioxidant defense and biosynthetic pathways. In the present report, we evaluated the effects of chemical compounds used as inhibitors of NNT over the last five decades, namely, 4-chloro-7-nitrobenzofurazan (NBD-Cl), N,N'-dicyclohexylcarbodiimide (DCC), palmitoyl-CoA, palmitoyl-l-carnitine, and rhein, on NNT activity and mitochondrial respiratory function. Concentrations of these compounds that partially inhibited the forward and reverse NNT reactions in detergent-solubilized mouse liver mitochondria significantly impaired mitochondrial respiratory function, as estimated by ADP-stimulated and nonphosphorylating respiration. Among the tested compounds, NBD-Cl showed the best relationship between NNT inhibition and low impact on respiratory function. Despite this, NBD-Cl concentrations that partially inhibited NNT activity impaired mitochondrial respiratory function and significantly decreased the viability of cultured Nnt-/- mouse astrocytes. We conclude that even though the tested compounds indeed presented inhibitory effects on NNT activity, at effective concentrations, they cause important undesirable effects on mitochondrial respiratory function and cell viability.


Subject(s)
Enzyme Inhibitors/pharmacology , Mitochondria, Liver/enzymology , NADP Transhydrogenase, AB-Specific/antagonists & inhibitors , NADP Transhydrogenase, AB-Specific/metabolism , Oxygen Consumption/drug effects , Animals , Enzyme Inhibitors/chemistry , Female , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , NADP Transhydrogenase, AB-Specific/genetics , Oxygen Consumption/genetics
14.
Amino Acids ; 52(2): 161-169, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31654209

ABSTRACT

Agmatine (AGM) produces a dual effect on the mitochondrial permeability transition (MPT) mechanism in rat liver mitochondria: at low concentrations, it induces the phenomenon, at high ones, inhibits it. The prevention at high concentrations is evidenced by the significant inhibition of mitochondrial swelling induced by Ca2+ and phosphate; in this condition, AGM both prevents the release of Apoptosis Inducing Factor (AIF) and enhances the release of other pro-apoptotic factors, such as cytochrome c (cyt c) and Smac/DIABLO. As these factors are released without MPT induction, the involvement of mitochondrial outer membrane permeabilization (MOMP) could be hypothesized. Cyclosporin A (CsA), a powerful inhibitor of MPT, enhanced the AGM-mediated inhibition of swelling, and surprisingly, prevented the release of cyt c and Smac/DIABLO. In the presence of Ca2+, AGM also activated the Bcl-2 family protein Bax, a key factor in inducing MOMP, which is inactivated by CsA. Together with the voltage-dependent anion channel (VDAC), Bax forms channels in the outer membrane further supporting the involvement of MOMP in the release of pro-apoptotic factors. In view of the fact that VDAC was inactivated by ruthenium red, which in turn inhibited the release of cyt c, it can be hypothesized that, on the one hand, AGM inhibits MPT induction and, on the other, it selectively permeabilizes the outer membrane via MOMP induction.


Subject(s)
Agmatine/metabolism , Apoptosis Inducing Factor/metabolism , Mitochondrial Membranes/metabolism , Animals , Apoptosis , Apoptosis Inducing Factor/genetics , Calcium/metabolism , Cell Membrane Permeability , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Rats
15.
Cell Biol Toxicol ; 36(6): 603-608, 2020 12.
Article in English | MEDLINE | ID: mdl-32607778

ABSTRACT

Aflatoxin B1 (AFB1) is widely distributed in nature and is confirmed to be the most toxic of all the aflatoxins, whose predominant metabolism site is the liver. As a well-studied and vital mode of epigenetic modifications, aberrant methylation of the promoters in eukaryotic cells may cause the silence of essential genes, affecting their related transcriptional pathways and ultimately leading to the development of disease and cancers. This study investigated the mechanisms of AFB1-induced hepatotoxicity in S phase-arrested L02 cells using single-cell RNA-seq and single-cell reduced representation bisulfite sequencing (RRBS). AFB1 induced apoptosis and cell cycle S phase arrest, reduced mitochondrial membrane potential (ΔΨm), and increased reactive oxygen species (ROS) generation, as well as the DNA methylation level. Hepatotoxicity mechanism patterns induced by AFB1 in S phase-arrested L02 cells were revealed by combining single-cell RNA-seq with single-cell RRBS analysis, in which DNA methylation played a role via regulating the gonadotropin-releasing hormone receptor pathway, the Wnt signaling pathway, and the TGF-beta signaling pathway. Moreover, a novel strategy for precision toxicology exploration was obtained, including the selection of target cells, multi-group non-directional sequencing, and pathway analysis.


Subject(s)
Aflatoxin B1/toxicity , DNA Methylation/drug effects , Gene Expression Profiling , Hepatocytes/drug effects , RNA-Seq , S Phase Cell Cycle Checkpoints/drug effects , Single-Cell Analysis , Transcriptome/drug effects , Apoptosis/drug effects , Cell Line , Gene Regulatory Networks/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Liver/drug effects , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism
16.
Int J Mol Sci ; 21(18)2020 Sep 19.
Article in English | MEDLINE | ID: mdl-32961796

ABSTRACT

MicroRNA-29a (miR-29a) has been shown to ameliorate hepatocellular damage, such as in the context of non-alcoholic fatty liver disease (NAFLD), steatohepatitis (NASH), and cholestatic injury. However, the mechanism mediating the hepatoprotective effect of miR-29a in diet-induced NASH remains elusive. In the present study, C57BL/6 mice of wild-type (WT) or miR-29a overexpression were fed with methionine-choline sufficient (MCS) or methionine-choline-deficient (MCD) diet for four weeks. The C57BL/6 mice harboring miR-29a overexpression presented reduced plasma AST, hepatic CD36, steatosis, and fibrosis induced by MCD. The TargetScan Release7.2-based bioinformatic analysis, KEGG pathway analysis, and luciferase reporter assay confirmed that miR-29a targets 3'UTR of glycogen synthase kinase 3 beta (Gsk3b) mRNA in the HepG2 hepatocyte cell line. Furthermore, miR-29a overexpression in the MCD-fed group resulted in inhibition of Gsk3b mRNA and GSK3ß protein levels in the liver. GSK3ß was notably expressed jointly with the extent of aggregated protein, which was then identified to be associated with mitochondrial unfolded protein response (UPRmt), but not with endoplasmic reticulum UPR (UPRER). Additionally, in silico analysis of protein-protein interaction, in vivo, and in vitro correlation analyses of protein expression demonstrated that GSK3ß closely associated with sirtuin 1(SIRT1). Finally, the implication of SIRT1-mediated mitochondrial biogenesis in the perturbation of proteostasis was observed. We herein provide novel insight into a hepatoprotective pathway, whereby miR-29a inhibits GSK3ß to repress SIRT1-mediated mitochondrial biogenesis, leading to alleviation of mitochondrial proteostatic stress and UPRmt in the context of NASH. miR-29a, GSK3ß, and SIRT1 could thus serve as possible therapeutic targets to improve the treatment of NAFLD/NASH.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , MicroRNAs/biosynthesis , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Proteostasis , Sirtuin 1/metabolism , Animals , Glycogen Synthase Kinase 3 beta/genetics , Mice , Mice, Transgenic , MicroRNAs/genetics , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Sirtuin 1/genetics , Unfolded Protein Response
17.
Int J Mol Sci ; 21(8)2020 Apr 17.
Article in English | MEDLINE | ID: mdl-32316419

ABSTRACT

Non-alcoholic steatohepatitis (NASH), a type of non-alcoholic fatty liver disease, is characterized as steatosis and inflammation in the liver. NLRP3 inflammasome activation is associated with NASH pathology. We hypothesized that suppressing the NLRP3 inflammasome could be effective in preventing NASH. We searched substances that could inhibit the activation of the NLRP3 inflammasome and identified sweroside as an NLRP3 inhibitor. We investigated whether sweroside can be applied to prevent the pathological symptoms associated with NASH in a methionine-choline-deficient (MCD) diet-induced NASH mouse model. The activation of the NLRP3 inflammasome was determined by detecting the production of caspase-1 and IL-1ß from pro-caspase-1 and pro-IL-1ß in primary mouse macrophages and mouse liver. In a NASH model, mice were fed an MCD diet for two weeks with daily intraperitoneal injections of sweroside. Sweroside effectively inhibited NLRP3 inflammasome activation in primary macrophages as shown by a decrease in IL-1ß and caspase-1 production. In a MCD diet-induced NASH mouse model, intraperitoneal injection of sweroside significantly reduced serum aspartate transaminase and alanine transaminase levels, hepatic immune cell infiltration, hepatic triglyceride accumulation, and liver fibrosis. The improvement of NASH symptoms by sweroside was accompanied with its inhibitory effects on the hepatic NLRP3 inflammasome as hepatic IL-1ß and caspase-1 were decreased. Furthermore, sweroside blocked de novo synthesis of mitochondrial DNA in the liver, contributing to suppression of the NLRP3 inflammasome. These results suggest that targeting the NLRP3 inflammasome with sweroside could be beneficially employed to improve NASH symptoms.


Subject(s)
Caspase 1/metabolism , Diet/adverse effects , Interleukin-1beta/metabolism , Iridoid Glucosides/administration & dosage , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Choline/metabolism , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Disease Models, Animal , Gene Expression Regulation , Injections, Intraperitoneal , Iridoid Glucosides/pharmacology , Liver/drug effects , Liver/metabolism , Macrophages/drug effects , Macrophages/metabolism , Methionine/deficiency , Mice , Mitochondria, Liver/drug effects , Mitochondria, Liver/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Treatment Outcome
18.
Am J Physiol Endocrinol Metab ; 317(4): E710-E722, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31430204

ABSTRACT

This study aimed to investigate whether hypoxia can affect nonalcoholic fatty liver disease (NAFLD) progression and the associated mechanisms, specifically regarding the hypoxia-inducible factor (HIF)-2α/peroxisome proliferator-activated receptor (PPAR)α pathway in vitro and in vivo. Recent studies have reported that, compared with HIF-1α, HIF-2α has different effects on lipid metabolism. We propose hypoxia may exacerbate NAFLD by the HIF-2α upregulation-induced suppression of PPARα in the liver. To verify this hypothesis, a steatotic human hepatocyte (L02) cell line treated with free fatty acids and a mouse model of NAFLD fed a high-fat diet were used. Steatotic hepatocytes were treated with hypoxia, HIF-2α siRNA, PPARα agonists, and inhibitors, respectively. Meanwhile, the NAFLD mice were exposed to intermittent hypoxia or intermittent hypoxia with PPARα agonists. The relative gene expression levels of HIF-1α, HIF-2α, mitochondrial function, fatty acid ß-oxidation and lipogenesis were examined. Evidence of lipid accumulation was observed, which demonstrated that, compared with normal hepatocytes, steatotic hepatocytes exhibited higher sensitivity to hypoxia. This phenomenon was closely associated with HIF-2α. Moreover, lipid accumulation in hepatocytes was ameliorated by HIF-2α silencing or a PPARα agonist, despite the hypoxia treatment. HIF-2α overexpression under hypoxic conditions suppressed PPARα, leading to PGC-1α, NRF-1, ESRRα downregulation, and mitochondrial impairment. Additionally, ß-oxidation genes such as CPT1α, CPT2α, ACOX1, and ACOX2 were downregulated and lipogenesis genes including LXRα, FAS, and SCD1 were upregulated by hypoxia. Therefore, we concluded that HIF-2α overexpression induced by hypoxia aggravated NAFLD progression by suppressing fatty acid ß-oxidation and inducing lipogenesis in the liver via PPARα.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia/genetics , Non-alcoholic Fatty Liver Disease/genetics , PPAR alpha/genetics , Signal Transduction/genetics , Animals , Cell Line , Diet, High-Fat , Hepatocytes/metabolism , Humans , Hypoxia/complications , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Mitochondria, Liver/genetics , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/complications , RNA, Small Interfering/pharmacology
19.
FASEB J ; 32(6): 3434-3447, 2018 06.
Article in English | MEDLINE | ID: mdl-29401633

ABSTRACT

Hydroxysteroid (17ß) dehydrogenases (HSD17Bs) form an enzyme family characterized by their ability to catalyze reactions in steroid and lipid metabolism. In the present study, we characterized the phenotype of HSD17B13-knockout (HSD17B13KO) mice deficient in Hsd17b13. In these studies, hepatic steatosis was detected in HSD17B13KO male mice, indicated by histologic analysis and by the increased triglyceride concentration in the liver, whereas reproductive performance and serum steroid concentrations were normal in HSD17B13KO mice. In line with these changes, the expression of key proteins in fatty acid synthesis, such as FAS, acetyl-CoA carboxylase 1, and SCD1, was increased in the HSD17B13KO liver. Furthermore, the knockout liver showed an increase in 2 acylcarnitines, suggesting impaired mitochondrial ß-oxidation in the presence of unaltered malonyl CoA and AMPK expression. The glucose tolerance did not differ between wild-type and HSD17B13KO mice in the presence of lower levels of glucose 6-phosphatase in HSD17B13KO liver compared with wild-type liver. Furthermore, microgranulomas and increased portal inflammation together with up-regulation of immune response genes were observed in HSD17B13KO mice. Our data indicate that disruption of Hsd17b13 impairs hepatic-lipid metabolism in mice, resulting in liver steatosis and inflammation, but the enzyme does not play a major role in the regulation of reproductive functions.-Adam, M., Heikelä, H., Sobolewski, C., Portius, D., Mäki-Jouppila, J., Mehmood, A., Adhikari, P., Esposito, I., Elo, L. L., Zhang, F.-P., Ruohonen, S. T., Strauss, L., Foti, M., Poutanen, M. Hydroxysteroid (17ß) dehydrogenase 13 deficiency triggers hepatic steatosis and inflammation in mice.


Subject(s)
17-Hydroxysteroid Dehydrogenases/deficiency , Fatty Liver/enzymology , Lipid Metabolism , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Fatty Liver/genetics , Fatty Liver/pathology , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Mice , Mice, Knockout , Mitochondria, Liver/enzymology , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Oxidation-Reduction , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
20.
FASEB J ; 32(2): 728-741, 2018 02.
Article in English | MEDLINE | ID: mdl-28974562

ABSTRACT

Age is an independent risk factor of multiple organ failure in patients with sepsis. However, the age-related mechanisms of injury are not known. AMPK is a crucial regulator of energy homeostasis, which controls mitochondrial biogenesis by activation of peroxisome proliferator-activated receptor-γ coactivator-α (PGC-1α) and disposal of defective organelles by autophagy. We investigated whether AMPK dysregulation might contribute to age-dependent liver injury in young (2-3 mo) and mature male mice (11-13 mo) subjected to sepsis. Liver damage was higher in mature mice than in young mice and was associated with impairment of hepatocyte mitochondrial function, structure, and biogenesis and reduced autophagy. At molecular analysis, there was a time-dependent nuclear translocation of the active phosphorylated catalytic subunits AMPKα1/α2 and PGC-1α in young, but not in mature, mice after sepsis. Treatment with the AMPK activator 5-amino-4-imidazolecarboxamide riboside-1-ß-d-ribofuranoside (AICAR) improved liver mitochondrial structure in both age groups compared with vehicle. In loss-of-function studies, young knockout mice with systemic deficiency of AMPKα1 exhibited greater liver injury than did wild-type mice after sepsis. Our study suggests that AMPK is important for liver metabolic recovery during sepsis. Although its function may diminish with age, pharmacological activation of AMPK may be of therapeutic benefit.-Inata, Y., Kikuchi, S., Samraj, R. S., Hake, P. W., O'Connor, M., Ledford, J. R., O'Connor, J., Lahni, P., Wolfe, V., Piraino, G., Zingarelli, B. Autophagy and mitochondrial biogenesis impairment contribute to age-dependent liver injury in experimental sepsis: dysregulation of AMP-activated protein kinase pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aging/metabolism , Autophagy , Cell Nucleus/enzymology , Liver/metabolism , Mitochondria, Liver/metabolism , Sepsis/metabolism , AMP-Activated Protein Kinases/genetics , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Aging/genetics , Aging/pathology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Cell Nucleus/genetics , Liver/injuries , Liver/pathology , Mice , Mice, Knockout , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Ribonucleotides/pharmacology , Sepsis/genetics , Sepsis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL