Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 67
1.
PLoS Biol ; 19(5): e3001252, 2021 05.
Article En | MEDLINE | ID: mdl-33983919

The mitochondrial ATP synthase emerges as key hub of cellular functions controlling the production of ATP, cellular signaling, and fate. It is regulated by the ATPase inhibitory factor 1 (IF1), which is highly abundant in neurons. Herein, we ablated or overexpressed IF1 in mouse neurons to show that IF1 dose defines the fraction of active/inactive enzyme in vivo, thereby controlling mitochondrial function and the production of mitochondrial reactive oxygen species (mtROS). Transcriptomic, proteomic, and metabolomic analyses indicate that IF1 dose regulates mitochondrial metabolism, synaptic function, and cognition. Ablation of IF1 impairs memory, whereas synaptic transmission and learning are enhanced by IF1 overexpression. Mechanistically, quenching the IF1-mediated increase in mtROS production in mice overexpressing IF1 reduces the increased synaptic transmission and obliterates the learning advantage afforded by the higher IF1 content. Overall, IF1 plays a key role in neuronal function by regulating the fraction of ATP synthase responsible for mitohormetic mtROS signaling.


Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Brain/metabolism , Cell Line , Hippocampus/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proton-Translocating ATPases/physiology , Primary Cell Culture , Proteins/physiology , Reactive Oxygen Species/metabolism , Signal Transduction , ATPase Inhibitory Protein
2.
Neurobiol Aging ; 98: 63-77, 2021 02.
Article En | MEDLINE | ID: mdl-33254080

Synaptic failure underlies cognitive impairment in Alzheimer's disease (AD). Cumulative evidence suggests a strong link between mitochondrial dysfunction and synaptic deficits in AD. We previously found that oligomycin-sensitivity-conferring protein (OSCP) dysfunction produces pronounced neuronal mitochondrial defects in AD brains and a mouse model of AD pathology (5xFAD mice). Here, we prevented OSCP dysfunction by overexpressing OSCP in 5xFAD mouse neurons in vivo (Thy-1 OSCP/5xFAD mice). This approach protected OSCP expression and reduced interaction of amyloid-beta (Aß) with membrane-bound OSCP. OSCP overexpression also alleviated F1Fo ATP synthase deregulation and preserved mitochondrial function. Moreover, OSCP modulation conferred resistance to Aß-mediated defects in axonal mitochondrial dynamics and motility. Consistent with preserved neuronal mitochondrial function, OSCP overexpression ameliorated synaptic injury in 5xFAD mice as demonstrated by preserved synaptic density, reduced complement-dependent synapse elimination, and improved synaptic transmission, leading to preserved spatial learning and memory. Taken together, our findings show the consequences of OSCP dysfunction in the development of synaptic stress in AD-related conditions and implicate OSCP modulation as a potential therapeutic strategy.


Alzheimer Disease/etiology , Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/physiology , Synaptic Transmission/genetics , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Gene Expression , Memory , Mice, Transgenic , Mitochondria/metabolism , Mitochondrial Dynamics/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Molecular Targeted Therapy , Neurons/metabolism , Spatial Learning
3.
Sci Rep ; 10(1): 17589, 2020 10 16.
Article En | MEDLINE | ID: mdl-33067541

Dendritic spine injury underlies synaptic failure in many neurological disorders. Mounting evidence suggests a mitochondrial pathway of local nonapoptotic caspase signaling in mediating spine pruning. However, it remains unclear whether this caspase signaling plays a key role in spine loss when severe mitochondrial functional defects are present. The answer to this question is critical especially for some pathological states, in which mitochondrial deficits are prominent and difficult to fix. F1Fo ATP synthase is a pivotal mitochondrial enzyme and the dysfunction of this enzyme involves in diseases with spinopathy. Here, we inhibited F1Fo ATP synthase function in primary cultured hippocampal neurons by using non-lethal oligomycin A treatment. Oligomycin A induced mitochondrial defects including collapsed mitochondrial membrane potential, dissipated ATP production, and elevated reactive oxygen species (ROS) production. In addition, dendritic mitochondria underwent increased fragmentation and reduced positioning to dendritic spines along with increased caspase 3 cleavage in dendritic shaft and spines in response to oligomycin A. Concurring with these dendritic mitochondrial changes, oligomycin A-insulted neurons displayed spine loss and altered spine architecture. Such oligomycin A-mediated changes in dendritic spines were substantially prevented by the inhibition of caspase activation by using a pan-caspase inhibitor, quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone (Q-VD-OPh). Of note, the administration of Q-VD-OPh showed no protective effect on oligomycin A-induced mitochondrial dysfunction. Our findings suggest a pivotal role of caspase 3 signaling in mediating spine injury and the modulation of caspase 3 activation may benefit neurons from spine loss in diseases, at least, in those with F1Fo ATP synthase defects.


Dendritic Spines/metabolism , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Death , Female , Hippocampus/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mitochondrial Proton-Translocating ATPases/physiology , Neurons/metabolism , Oligomycins/metabolism , Oligomycins/pharmacology , Quinolines/pharmacology , Reactive Oxygen Species/metabolism
4.
J Bioenerg Biomembr ; 51(5): 315-328, 2019 10.
Article En | MEDLINE | ID: mdl-31418131

The yeast mitochondrial ATP synthase is a rotary molecular machine primarily responsible for the production of energy used to drive cellular processes. The enzyme complex is composed of 17 different subunits grouped into a soluble F1 sector and a membrane-embedded F0 sector. The catalytic head of the F1 sector and the membrane integrated motor module in the F0 sector are connected by two stalks, the F1 central stalk and the F0 peripheral stalk. Proton translocation through the F0 motor module drives the rotation of the subunit 910-ring that generates torque which is transmitted to the calaytic head through the γ subunit of the central stalk. The rotation of the γ subunit causes changes in conformation of the catalytic head which leads to the synthesis of ATP. Biogenesis of the enzyme involves modular assembly of polypeptides of dual genetic origin, the nuclear and the mitochondrial genomes. Most of the yeast ATP synthase subunits are encoded by the genome of the nucleus, translated on cytosolic ribosomes and imported into mitochondria. In the mitochondria, the enzyme forms a dimer which contributes to the formation of cristae, a characteristic of mitochondrial morphology. Substantial progress has recently been made on the elucidation of detailed stucture, function and biogenesis of yeast mitochondrial ATP synthase. The recent availability of high-resolution structure of the complete monomeric form, as well as the atomic model for the dimeric F0 sector, has advanced the understanding of the enzyme complex. This review is intended to provide an overview of current understanding of the molecular structure, catalytic mechanism, subunit import into mitochondria, and the subunit assembly into the enzyme complex. This is important as the yeast mitochondrial ATP synthase may be used as a model for understanding the corresponding enzyme complexes from human and other eukaryotic cells in physiological and diseased states.


Fungal Proteins , Mitochondrial Proton-Translocating ATPases , Biological Transport , Catalysis , Fungal Proteins/biosynthesis , Fungal Proteins/chemistry , Fungal Proteins/physiology , Mitochondrial Proton-Translocating ATPases/biosynthesis , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/physiology , Models, Molecular , Molecular Structure , Protein Subunits/metabolism
5.
J Proteomics ; 175: 136-143, 2018 03 20.
Article En | MEDLINE | ID: mdl-29325989

Apostichopus japonicus is one of the most important aquaculture species in China. Skin ulceration syndrome (SUS) of sea cucumber is a common and serious disease affected the development of A. japonicus culture industry. To better understand the response mechanisms of A. japonicus during SUS progression, the protein variations in the body wall of A. japonicus at different stages of SUS were investigated by a comparative proteomic approach based on isobaric tags for relative and absolute quantification. A total of 1449 proteins were identified from the samples at different SUS stages. Among these proteins, 145 proteins were differentially expressed in the SUS-related samples compared to those of healthy A. japonicus. These differentially expressed proteins involved a wide range of functions. Among these differentially expressed proteins, only two proteins, alpha-5-collagen and an unknown function protein, were differentially expressed during the whole progression of SUS compared with healthy A. japonicus. In addition, ATP synthase subunit beta (ATP5ß) interacted with a variety of proteins with different functions during the SUS progression. These results implied that alpha-5-collagen and ATP5ß could play important roles during the SUS progression of A. japonicus. Our study provided a new sight to understand the molecular responses of sea cucumber during the SUS progression and accumulated data for the prevention of SUS in sea cucumber aquaculture. BIOLOGICAL SIGNIFICANCE: The current study aimed to reveal how the body wall of Apostichopus japonicus response to skin ulceration syndrome (SUS). To the best of our knowledge, this is the first proteomic study analyzing the differences in protein profile of sea cucumber during the whole SUS progression. By analyzing the expression differences of the proteome via isobaric labeling-based quantitative proteomic, we identified some proteins which may play important roles during the SUS progression. According to the enrichment analyses of these proteins based on Gene Ontology and Kyoto Encyclopedia of Genes and Genomes, a draft view of how the sea cucumber affected by SUS has been drawn. The common and unique differentially expressed proteins by Venn analysis showed that alpha-5-collagen was down-regulated at all stages of SUS, which had the potential as a target component for the host-directed SUS therapy. In addition, ATP5ß, a subunit of mitochondrial ATP synthase, interacting with a variety of proteins with different functions during the SUS progression. This result illustrated that energy production and metabolism could play an important role in the formation of skin ulceration and resistance to pathogens in sea cucumber. The results of this study will be helpful for researchers to gain insights into the complex molecular mechanism of SUS in sea cucumber.


Collagen/physiology , Mitochondrial Proton-Translocating ATPases/physiology , Proteomics/methods , Skin Ulcer/pathology , Animals , Disease Progression , Energy Metabolism , Gene Expression Profiling , Sea Cucumbers , Skin Ulcer/genetics , Syndrome
6.
Sci Rep ; 7(1): 17417, 2017 12 12.
Article En | MEDLINE | ID: mdl-29234032

The pathogenesis of heart failure associated with dilated cardiomyopathy (DCM) may result in part from adenosine triphosphate (ATP) dysregulation in the myocardium. Under these conditions, diabetes-associated protein in insulin-sensitive tissue (DAPIT), which is encoded by the upregulated during skeletal muscle growth 5 (USMG5) gene, plays a crucial role in energy production by mitochondrial ATP synthase. To determine whether USMG5 is related to the development of heart failure, we performed clinical and experimental studies. Microarray analysis showed that the expression levels of USMG5 were positively correlated with those of natriuretic peptide precursor A in the human failed myocardium. When endogenous z-usmg5 in zebrafish was disrupted using morpholino (MO) oligonucleotides, the pericardial sac and atrial areas were larger and ventricular fractional shortening was reduced compared to in the control MO group. The expression levels of natriuretic peptides were upregulated in the z-usmg5 MO group compared to in controls. Further, microarray analysis revealed that genes in the calcium signalling pathway were downregulated in the z-usmg5 MO group. These results demonstrate that DAPIT plays a crucial role in the development of heart failure associated with DCM and thus may be a therapeutic target for heart failure.


Cardiomyopathy, Dilated/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Zebrafish Proteins/physiology , Animals , Atrial Natriuretic Factor/metabolism , Cardiomyopathy, Dilated/pathology , Gene Expression , Gene Knockdown Techniques , Heart Failure/metabolism , Heart Failure/pathology , Humans , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/physiology , Myocardium/metabolism , Myocardium/pathology , Zebrafish , Zebrafish Proteins/genetics
7.
Handb Exp Pharmacol ; 240: 21-46, 2017.
Article En | MEDLINE | ID: mdl-27590224

Mitochondrial ATP generation by oxidative phosphorylation combines the stepwise oxidation by the electron transport chain (ETC) of the reducing equivalents NADH and FADH2 with the generation of ATP by the ATP synthase. Recent studies show that the ATP synthase is not only essential for the generation of ATP but may also contribute to the formation of the mitochondrial permeability transition pore (PTP). We present a model, in which the PTP is located within the c-subunit ring in the Fo subunit of the ATP synthase. Opening of the PTP was long associated with uncoupling of the ETC and the initiation of programmed cell death. More recently, it was shown that PTP opening may serve a physiologic role: it can transiently open to regulate mitochondrial signaling in mature cells, and it is open in the embryonic mouse heart. This review will discuss how the ATP synthase paradoxically lies at the center of both ATP generation and cell death.


Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Proton-Translocating ATPases/physiology , Adenosine Triphosphate/biosynthesis , Animals , Apoptosis , Electron Transport , Energy Metabolism , Humans , Mitochondrial Permeability Transition Pore
8.
Aging Male ; 19(3): 187-191, 2016 Sep.
Article En | MEDLINE | ID: mdl-27187822

Mitochondrial defects have been related to obesity and prostate cancer. We investigated if Mexican-Mestizo men presenting this type of cancer, exhibited somatic mutations of ATP6 and/or ND3.Body mass index (BMI) was determined; the degree of prostate cancer aggressiveness was demarcated by the Gleason score. DNA from tumor tissue and from blood leukocytes was amplified by the polymerase chain reaction and ATP6 and ND3 were sequenced. We included 77 men: 20 had normal BMI, 38 were overweight and 19 had obesity; ages ranged from 52 to 83. After sequencing ATP6 and ND3, from DNA obtained from leukocytes and tumor tissue, we did not find any somatic mutations. All changes observed, in both genes, were polymorphisms. In ATP6 we identified, in six patients, two non-synonymous nucleotide changes and in ND3 we observed that twelve patients presented non-synonymous polymorphisms. To our knowledge, this constitutes the first report where the complete sequences of the ATP6 and ND3 have been analyzed in Mexican-Mestizo men with prostate cancer and diverse BMI. Our results differ with those reported in Caucasian populations, possibly due to ethnic differences.


Electron Transport Complex I/physiology , Mitochondrial Proton-Translocating ATPases/physiology , Obesity/genetics , Overweight/genetics , Polymorphism, Genetic , Prostatic Neoplasms/genetics , Aged , Aged, 80 and over , Electron Transport Complex I/genetics , Humans , Male , Mexico , Middle Aged , Mitochondrial Proton-Translocating ATPases/genetics , Neoplasm Metastasis/genetics , Obesity/complications , Overweight/complications , Prostatic Neoplasms/complications , Prostatic Neoplasms/pathology
9.
Oxid Med Cell Longev ; 2016: 3869610, 2016.
Article En | MEDLINE | ID: mdl-27034734

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) targeting mitochondria are major causative factors in disease pathogenesis. The mitochondrial permeability transition pore (PTP) is a mega-channel modulated by calcium and ROS/RNS modifications and it has been described to play a crucial role in many pathophysiological events since prolonged channel opening causes cell death. The recent identification that dimers of ATP synthase form the PTP and the fact that posttranslational modifications caused by ROS/RNS also affect cellular bioenergetics through the modulation of ATP synthase catalysis reveal a dual function of these modifications in the cells. Here, we describe mitochondria as a major site of production and as a target of ROS/RNS and discuss the pathophysiological conditions in which oxidative and nitrosative modifications modulate the catalytic and pore-forming activities of ATP synthase.


Cell Death , Energy Metabolism , Mitochondrial Proton-Translocating ATPases/physiology , Reactive Nitrogen Species/physiology , Reactive Oxygen Species/pharmacology , Animals , Cell Death/drug effects , Energy Metabolism/drug effects , Humans , Mitochondria/metabolism , Oxidation-Reduction , Reactive Nitrogen Species/pharmacology , Reactive Oxygen Species/metabolism
10.
Biochim Biophys Acta ; 1847(11): 1347-53, 2015 Nov.
Article En | MEDLINE | ID: mdl-26050973

The free radical theory of aging is almost 60 years old. As mitochondria are the principle source of intracellular reactive oxygen species (ROS), this hypothesis suggested a central role for the mitochondrion in normal mammalian aging. In recent years, however, much work has questioned the importance of mitochondrial ROS in driving aging. Conversely new evidence points to other facets of mitochondrial dysfunction which may nevertheless suggest the mitochondrion retains a critical role at the center of a complex web of processes leading to cellular and organismal aging.


Aging/physiology , Mitochondria/physiology , Animals , DNA, Mitochondrial/genetics , Humans , Mitochondrial Proton-Translocating ATPases/physiology , Mutation , Reactive Oxygen Species/metabolism
11.
Biochim Biophys Acta ; 1847(9): 849-60, 2015 Sep.
Article En | MEDLINE | ID: mdl-25667968

F1F0-ATP synthases are multimeric protein complexes and common prerequisites for their correct assembly are (i) provision of subunits in appropriate relative amounts, (ii) coordination of membrane insertion and (iii) avoidance of assembly intermediates that uncouple the proton gradient or wastefully hydrolyse ATP. Accessory factors facilitate these goals and assembly occurs in a modular fashion. Subcomplexes common to bacteria and mitochondria, but in part still elusive in chloroplasts, include a soluble F1 intermediate, a membrane-intrinsic, oligomeric c-ring, and a membrane-embedded subcomplex composed of stator subunits and subunit a. The final assembly step is thought to involve association of the preformed F1-c10-14 with the ab2 module (or the ab8-stator module in mitochondria)--mediated by binding of subunit δ in bacteria or OSCP in mitochondria, respectively. Despite the common evolutionary origin of F1F0-ATP synthases, the set of auxiliary factors required for their assembly in bacteria, mitochondria and chloroplasts shows clear signs of evolutionary divergence. This article is part of a Special Issue entitled: Chloroplast Biogenesis.


Mitochondrial Proton-Translocating ATPases/chemistry , Chloroplasts/enzymology , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/physiology
12.
Biochem J ; 466(3): 601-11, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25588698

Mutations in the MT-ATP6 gene are frequent causes of severe mitochondrial disorders. Typically, these are missense mutations, but another type is represented by the 9205delTA microdeletion, which removes the stop codon of the MT-ATP6 gene and affects the cleavage site in the MT-ATP8/MT-ATP6/MT-CO3 polycistronic transcript. This interferes with the processing of mRNAs for the Atp6 (Fo-a) subunit of ATP synthase and the Cox3 subunit of cytochrome c oxidase (COX). Two cases described so far presented with strikingly different clinical phenotypes-mild transient lactic acidosis or fatal encephalopathy. To gain more insight into the pathogenic mechanism, we prepared 9205delTA cybrids with mutation load ranging between 52 and 99% and investigated changes in the structure and function of ATP synthase and the COX. We found that 9205delTA mutation strongly reduces the levels of both Fo-a and Cox3 proteins. Lack of Fo-a alters the structure but not the content of ATP synthase, which assembles into a labile, ∼60 kDa smaller, complex retaining ATP hydrolytic activity but which is unable to synthesize ATP. In contrast, lack of Cox3 limits the biosynthesis of COX but does not alter the structure of the enzyme. Consequently, the diminished mitochondrial content of COX and non-functional ATP synthase prevent most mitochondrial ATP production. The biochemical effects caused by the 9205delTA microdeletion displayed a pronounced threshold effect above ∼90% mutation heteroplasmy. We observed a linear relationship between the decrease in subunit Fo-a or Cox3 content and the functional presentation of the defect. Therefore we conclude that the threshold effect originated from a gene-protein level.


DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , Mitochondrial Proton-Translocating ATPases/physiology , Mutation/genetics , Cell Line , Electron Transport Complex IV/metabolism , Gene Deletion , Humans , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/deficiency , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/metabolism
13.
FASEB J ; 29(5): 1676-87, 2015 May.
Article En | MEDLINE | ID: mdl-25526730

The importance of epigenetic changes in the development of hepatic steatosis is largely unknown. The histone variant macroH2A1 under alternative splicing gives rise to macroH2A1.1 and macroH2A1.2. In this study, we show that the macroH2A1 isoforms play an important role in the regulation of lipid accumulation in hepatocytes. Hepatoma cell line and immortalized human hepatocytes transiently transfected or knocked down with macroH2A1 isoforms were used as in vitro model of fat-induced steatosis. Gene expressions were analyzed by quantitative PCR array and Western blot. Chromatin immunoprecipitation analysis was performed to check the association of histone H3 lysine 27 trimethylation (H3K27me3) and histone H3 lysine 4 trimethylation (H3K4me3) with the promoter of lipogenic genes. Livers from knockout mice that are resistant to lipid deposition despite a high-fat diet were used for histopathology. We found that macroH2A1.2 is regulated by fat uptake and that its overexpression caused an increase in lipid uptake, triglycerides, and lipogenic genes compared with macroH2A1.1. This suggests that macroH2A1.2 is important for lipid uptake, whereas macroH2A1.1 was found to be protective. The result was supported by a high positivity for macroH2A1.1 in knockout mice for genes targeted by macroH2A1 (Atp5a1 and Fam73b), that under a high-fat diet presented minimal lipidosis. Moreover, macroH2A1 isoforms differentially regulate the expression of lipogenic genes by modulating the association of the active (H3K4me3) and repressive (H3K27me3) histone marks on their promoters. This study underlines the importance of the replacement of noncanonical histones in the regulation of genes involved in lipid metabolism in the progression of steatosis.


Biomarkers/metabolism , Carcinoma, Hepatocellular/pathology , Diet, High-Fat/adverse effects , Epigenomics , Fatty Liver/metabolism , Fatty Liver/pathology , Hepatocytes/pathology , Histones/metabolism , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Fatty Liver/etiology , Female , Fluorescent Antibody Technique , Gene Expression Profiling , Hepatocytes/metabolism , Histones/genetics , Humans , Immunoenzyme Techniques , Lipid Peroxidation , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proton-Translocating ATPases/physiology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Protein Isoforms , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
14.
Cell Calcium ; 56(6): 437-45, 2014 Dec.
Article En | MEDLINE | ID: mdl-25454774

This review covers recent progress on the nature of the mitochondrial permeability transition pore (PTP) - a key effector in the mitochondrial pathways to cell death - and on the adaptive responses of tumor cells that desensitize the PTP to Ca(2+) and reactive oxygen species (ROS), thereby playing an important role in the resistance of tumors to cell death. The discovery that the PTP forms from dimers of F-ATP synthase; and the definition of the Ca(2+)- and ROS-dependent signaling pathways affecting the transition of the F-ATP synthase from an energy-conserving to an energy-dissipating device open new perspectives for therapeutic intervention in cancer cells.


Adaptation, Physiological/physiology , Mitochondrial Membrane Transport Proteins/physiology , Neoplasms/pathology , Neoplasms/physiopathology , Animals , Apoptosis/physiology , Calcium/metabolism , Humans , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/physiology , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
15.
Biochim Biophys Acta ; 1837(6): 940-52, 2014 Jun.
Article En | MEDLINE | ID: mdl-24650628

Archaea live under different environmental conditions, such as high salinity, extreme pHs and cold or hot temperatures. How energy is conserved under such harsh environmental conditions is a major question in cellular bioenergetics of archaea. The key enzymes in energy conservation are the archaeal A1AO ATP synthases, a class of ATP synthases distinct from the F1FO ATP synthase ATP synthase found in bacteria, mitochondria and chloroplasts and the V1VO ATPases of eukaryotes. A1AO ATP synthases have distinct structural features such as a collar-like structure, an extended central stalk, and two peripheral stalks possibly stabilizing the A1AO ATP synthase during rotation in ATP synthesis/hydrolysis at high temperatures as well as to provide the storage of transient elastic energy during ion-pumping and ATP synthesis/-hydrolysis. High resolution structures of individual subunits and subcomplexes have been obtained in recent years that shed new light on the function and mechanism of this unique class of ATP synthases. An outstanding feature of archaeal A1AO ATP synthases is their diversity in size of rotor subunits and the coupling ion used for ATP synthesis with H(+), Na(+) or even H(+) and Na(+) using enzymes. The evolution of the H(+) binding site to a Na(+) binding site and its implications for the energy metabolism and physiology of the cell are discussed.


Archaea/enzymology , Archaeal Proteins/physiology , Mitochondrial Proton-Translocating ATPases/physiology , Molecular Motor Proteins/physiology , Archaeal Proteins/chemistry , Binding Sites , Biocatalysis , Catalytic Domain , Mitochondrial Proton-Translocating ATPases/chemistry , Models, Molecular , Molecular Motor Proteins/chemistry
16.
Diabetes ; 63(3): 947-59, 2014 Mar.
Article En | MEDLINE | ID: mdl-24296716

ATP synthase ß subunit (ATPSß) had been previously shown to play an important role in controlling ATP synthesis in pancreatic ß-cells. This study aimed to investigate the role of ATPSß in regulation of hepatic ATP content and glucose metabolism in diabetic mice. ATPSß expression and ATP content were both reduced in the livers of type 1 and type 2 diabetic mice. Hepatic overexpression of ATPSß elevated cellular ATP content and ameliorated hyperglycemia of streptozocin-induced diabetic mice and db/db mice. ATPSß overexpression increased phosphorylated Akt (pAkt) levels and reduced PEPCK and G6pase expression levels in the livers. Consistently, ATPSß overexpression repressed hepatic glucose production in db/db mice. In cultured hepatocytes, ATPSß overexpression increased intracellular and extracellular ATP content, elevated the cytosolic free calcium level, and activated Akt independent of insulin. The ATPSß-induced increase in cytosolic free calcium and pAkt levels was attenuated by inhibition of P2 receptors. Notably, inhibition of calmodulin (CaM) completely abolished ATPSß-induced Akt activation in liver cells. Inhibition of P2 receptors or CaM blocked ATPSß-induced nuclear exclusion of forkhead box O1 in liver cells. In conclusion, a decrease in hepatic ATPSß expression in the liver, leading to the attenuation of ATP-P2 receptor-CaM-Akt pathway, may play an important role in the progression of diabetes.


Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/prevention & control , Liver/metabolism , Mitochondrial Proton-Translocating ATPases/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Animals , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gluconeogenesis , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Transport , Signal Transduction , Streptozocin
17.
Zhongguo Dang Dai Er Ke Za Zhi ; 15(7): 596-600, 2013 Jul.
Article Zh | MEDLINE | ID: mdl-23866288

The mammalian mitochondrial ATP synthase, also as known as mitochondrial respiratory chain complex V, is a large protein complex located in the mitochondrial inner membrane, where it catalyzes ATP synthesis from ADP, Pi, and Mg2+ at the expense of an electrochemical gradient of protons generated by the electron transport chain. Complex V is composed of 2 functional domains F0 and F1. The clinical features of patients are significantly heterogeneous depending on the involved organs. Most patients with complex V deficiency had clinical onset in the neonatal period with severe brain damage or multi-organ failure resulting in a high mortality. Neuromuscular disorders, cardiomyopathy, lactic acidosis and 3-methylglutaconic aciduria are common findings. Complex V consists of 16 subunits encoded by both mitochondrial DNA and nuclear DNA. On MT-ATP6, MT-ATP8, ATPAF2, TMEM70 and ATP5E gene of mitochondrial DNA, many mutations associated with Complex V deficiency have been identified. Here, the pathology, clinical features, diagnosis, treatment and molecular genetics of Complex V deficiency were summarized.


Mitochondrial Diseases/complications , Mitochondrial Diseases/etiology , Mitochondrial Proton-Translocating ATPases/deficiency , Mitochondrial Diseases/therapy , Mitochondrial Proton-Translocating ATPases/chemistry , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/physiology , Prognosis
18.
Blood ; 120(6): 1246-53, 2012 Aug 09.
Article En | MEDLINE | ID: mdl-22753871

Antigen-presenting cells (APCs) act as vehicles that transfer HIV to their target CD4(+) cells through an intercellular junction, termed the virologic synapse. The molecules that are involved in this process remain largely unidentified. In this study, we used photoaffinity labeling and a proteomic approach to identify new proteins that facilitate HIV-1 transfer. We identified ectopic mitochondrial ATP synthase as a factor that mediates HIV-1 transfer between APCs and CD4(+) target cells. Monoclonal antibodies against the ß-subunit of ATP synthase inhibited APC-mediated transfer of multiple strains HIV-1 to CD4(+) target cells. Likewise, the specific inhibitors of ATPase, citreoviridin and IF1, completely blocked APC-mediated transfer of HIV-1 at the APC-target cell interaction step. Confocal fluorescent microscopy showed localization of extracellular ATP synthase at junctions between APC and CD4(+) target cells. We conclude that ectopic ATP synthase could be an accessible molecular target for inhibiting HIV-1 proliferation in vivo.


Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/metabolism , HIV-1/metabolism , Mitochondrial Proton-Translocating ATPases/physiology , Antibodies/pharmacology , Antigen Presentation/physiology , Antigen-Presenting Cells/immunology , Biological Transport/immunology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , HEK293 Cells , HIV-1/immunology , HeLa Cells , Humans , Intercellular Junctions/immunology , Intercellular Junctions/metabolism , Mitochondrial Proton-Translocating ATPases/immunology , Mitochondrial Proton-Translocating ATPases/metabolism , Photoaffinity Labels/pharmacology , Protein Transport/immunology , Staining and Labeling/methods , Tissue Distribution
19.
Comp Biochem Physiol B Biochem Mol Biol ; 162(4): 107-12, 2012 Aug.
Article En | MEDLINE | ID: mdl-22507548

The effect of hypoxia and re-oxygenation on the mitochondrial complex F(O)F(1)-ATP synthase was investigated in the whiteleg shrimp Litopenaeus vannamei. A 660 kDa protein complex isolated from mitochondria of the shrimp muscle was identified as the ATP synthase complex. After 10h at hypoxia (1.5-2.0 mg oxygen/L), the concentration of L-lactate in plasma increased significantly, but the ATP amount and the concentration of ATPß protein remained unaffected. Nevertheless, an increase of 70% in the ATPase activity was detected, suggesting that the enzyme may be regulated at a post-translational level. Thus, during hypoxia shrimp are able to maintain ATP amounts probably by using some other energy sources as phosphoarginine when an acute lack of energy occurs. During re-oxygenation, the ATPase activity decreased significantly and the ATP production continued via the electron transport chain and oxidative phosphorylation. The results obtained showed that shrimp faces hypoxia partially by hydrolyzing the ATP through the reaction catalyzed by the mitochondrial ATPase which increases its activity.


Arthropod Proteins/physiology , Mitochondria, Muscle/enzymology , Mitochondrial Proton-Translocating ATPases/physiology , Muscles/enzymology , Penaeidae/metabolism , Adenosine Triphosphate/metabolism , Anaerobiosis , Animals , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Cell Hypoxia , Gene Expression , Lactic Acid/blood , Mitochondrial Proton-Translocating ATPases/metabolism , Muscles/cytology , Muscles/physiology , Oxygen/blood , Protein Subunits/genetics , Protein Subunits/metabolism , Tail
...