Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
Cell Rep ; 35(2): 108983, 2021 04 13.
Article En | MEDLINE | ID: mdl-33852870

Preclinical models of ischemia/reperfusion injury (RI) demonstrate the deleterious effects of permeability transition pore complex (PTPC) opening in the first minutes upon revascularization of the occluded vessel. The ATP synthase c subunit (Csub) influences PTPC activity in cells, thus impacting tissue injury. A conserved glycine-rich domain in Csub is classified as critical because, when mutated, it modifies ATP synthase properties, protein interaction with the mitochondrial calcium (Ca2+) uniporter complex, and the conductance of the PTPC. Here, we document the role of a naturally occurring mutation in the Csub-encoding ATP5G1 gene at the G87 position found in two ST-segment elevation myocardial infarction (STEMI) patients and how PTPC opening is related to RI in patients affected by the same disease. We report a link between the expression of ATP5G1G87E and the response to hypoxia/reoxygenation of human cardiomyocytes, which worsen when compared to those expressing the wild-type protein, and a positive correlation between PTPC and RI.


Hypoxia/genetics , Mitochondria/genetics , Mitochondrial Permeability Transition Pore/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Myocytes, Cardiac/metabolism , ST Elevation Myocardial Infarction/genetics , Aged , Animals , Base Sequence , Calcium Channels/genetics , Calcium Channels/metabolism , Exons , Female , Gene Expression , Humans , Hypoxia/metabolism , Hypoxia/pathology , Introns , Male , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proton-Translocating ATPases/deficiency , Mutation , Myocytes, Cardiac/pathology , Oxygen/adverse effects , Prospective Studies , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , ST Elevation Myocardial Infarction/metabolism , ST Elevation Myocardial Infarction/pathology
2.
Int J Mol Sci ; 21(3)2020 Feb 06.
Article En | MEDLINE | ID: mdl-32041178

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is an enigmatic condition characterized by exacerbation of symptoms after exertion (post-exertional malaise or "PEM"), and by fatigue whose severity and associated requirement for rest are excessive and disproportionate to the fatigue-inducing activity. There is no definitive molecular marker or known underlying pathological mechanism for the condition. Increasing evidence for aberrant energy metabolism suggests a role for mitochondrial dysfunction in ME/CFS. Our objective was therefore to measure mitochondrial function and cellular stress sensing in actively metabolizing patient blood cells. We immortalized lymphoblasts isolated from 51 ME/CFS patients diagnosed according to the Canadian Consensus Criteria and an age- and gender-matched control group. Parameters of mitochondrial function and energy stress sensing were assessed by Seahorse extracellular flux analysis, proteomics, and an array of additional biochemical assays. As a proportion of the basal oxygen consumption rate (OCR), the rate of ATP synthesis by Complex V was significantly reduced in ME/CFS lymphoblasts, while significant elevations were observed in Complex I OCR, maximum OCR, spare respiratory capacity, nonmitochondrial OCR and "proton leak" as a proportion of the basal OCR. This was accompanied by a reduction of mitochondrial membrane potential, chronically hyperactivated TOR Complex I stress signaling and upregulated expression of mitochondrial respiratory complexes, fatty acid transporters, and enzymes of the ß-oxidation and TCA cycles. By contrast, mitochondrial mass and genome copy number, as well as glycolytic rates and steady state ATP levels were unchanged. Our results suggest a model in which ME/CFS lymphoblasts have a Complex V defect accompanied by compensatory upregulation of their respiratory capacity that includes the mitochondrial respiratory complexes, membrane transporters and enzymes involved in fatty acid ß-oxidation. This homeostatically returns ATP synthesis and steady state levels to "normal" in the resting cells, but may leave them unable to adequately respond to acute increases in energy demand as the relevant homeostatic pathways are already activated.


Adenosine Triphosphate/metabolism , Fatigue Syndrome, Chronic/metabolism , Lymphocytes/cytology , Mitochondrial Proton-Translocating ATPases/deficiency , Adult , Aged , Canada , Cell Culture Techniques , Cell Proliferation , Cell Survival , Cells, Cultured , Energy Metabolism , Female , Humans , Lymphocytes/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Middle Aged , Mitochondria/metabolism , Oxygen Consumption , Proteomics/methods
3.
Biochem Biophys Res Commun ; 521(4): 1036-1041, 2020 01 22.
Article En | MEDLINE | ID: mdl-31732150

Mitochondrial ATP synthase is responsible for production of the majority of cellular ATP. Disorders of ATP synthase in humans can be caused by numerous mutations in both structural subunits and specific assembly factors. They are associated with variable pathogenicity and clinical phenotypes ranging from mild to the most severe mitochondrial diseases. To shed light on primary/pivotal functional consequences of ATP synthase deficiency, we explored human HEK 293 cells with a varying content of fully assembled ATP synthase, selectively downregulated to 15-80% of controls by the knockdown of F1 subunits γ, δ and ε. Examination of cellular respiration and glycolytic flux revealed that enhanced glycolysis compensates for insufficient mitochondrial ATP production while reduced dissipation of mitochondrial membrane potential leads to elevated ROS production. Both insufficient energy provision and increased oxidative stress contribute to the resulting pathological phenotype. The threshold for manifestation of the ATP synthase defect and subsequent metabolic remodelling equals to 10-30% of residual ATP synthase activity. The metabolic adaptations are not able to sustain proliferation in a galactose medium, although sufficient under glucose-rich conditions. As metabolic alterations occur when the content of ATP synthase drops below 30%, some milder ATP synthase defects may not necessarily manifest with a mitochondrial disease phenotype, as long as the threshold level is not exceeded.


Mitochondrial Proton-Translocating ATPases/deficiency , Cell Survival , Clone Cells , Gene Knockdown Techniques , Glycolysis , HEK293 Cells , Humans , Inhibitory Concentration 50 , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Stress , Thermodynamics
4.
Proc Natl Acad Sci U S A ; 116(26): 12816-12821, 2019 06 25.
Article En | MEDLINE | ID: mdl-31213546

The opening of the permeability transition pore, a nonspecific channel in inner mitochondrial membranes, is triggered by an elevated total concentration of calcium ions in the mitochondrial matrix, leading to disruption of the inner membrane and necrotic cell death. Cyclosporin A inhibits pore opening by binding to cyclophilin D, which interacts with the pore. It has been proposed that the pore is associated with the ATP synthase complex. Previously, we confirmed an earlier observation that the pore survives in cells lacking membrane subunits ATP6 and ATP8 of ATP synthase, and in other cells lacking the enzyme's c8 rotor ring or, separately, its peripheral stalk subunits b and oligomycin sensitive conferral protein. Here, we investigated whether the pore is associated with the remaining membrane subunits of the enzyme. Individual deletion of subunits e, f, g, and 6.8-kDa proteolipid disrupts dimerization of the complex, and deletion of DAPIT (diabetes-associated protein in insulin sensitive tissue) possibly influences oligomerization of dimers, but removal of each subunit had no effect on the pore. Also, we removed together the enzyme's membrane bound c8 ring and the δ-subunit from the catalytic domain. The resulting cells assemble only a subcomplex derived from the peripheral stalk and membrane-associated proteins. Despite diminished levels of respiratory complexes, these cells generate a membrane potential to support uptake of calcium into the mitochondria, leading to pore opening, and retention of its characteristic properties. It is most unlikely that the ATP synthase, dimer or monomer, or any component, provides the permeability transition pore.


Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/deficiency , Cell Line , Humans , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Protein Multimerization
5.
Pest Manag Sci ; 75(5): 1425-1434, 2019 May.
Article En | MEDLINE | ID: mdl-30417535

BACKGROUND: RNA interference (RNAi) techniques have emerged as powerful tools to develop novel management strategies for the control of insect pests. The leafhopper Euscelidius variegatus is a natural vector of chrysanthemum yellows phytoplasma and a laboratory vector of Flavescence dorée phytoplasma. Phytoplasmas are insect-borne bacterial plant pathogens that cause economically relevant crop losses worldwide. RESULTS: In this study, we demonstrated that microinjection of muscle actin and ATP synthase ß double-stranded (ds)RNAs into adult insects caused an exponential reduction in the expression of both genes, which began within 72 h of dsRNA administration and lasted for 14 days, leading to almost complete silencing of the target genes. Such silencing effects on muscle actin expression appeared to be both time- and dose-dependent. Our results also showed that the knockdown of both genes caused a significant decrease in survival rates in comparison with green fluorescent protein (GFP) dsRNA-injected control insects. CONCLUSION: The effectiveness of RNAi-based gene silencing in E. variegatus guarantees the availability of a powerful reverse genetic tool for the functional annotation of its genes and the identification of those potentially involved in the interaction with phytoplasmas. In addition, this study demonstrated that muscle actin and ATP synthase ß may represent candidate genes for RNAi-based control of E. variegatus. © 2018 Society of Chemical Industry.


Actins/genetics , Hemiptera/genetics , Hemiptera/physiology , Mitochondrial Proton-Translocating ATPases/genetics , Muscles/metabolism , Phytoplasma/physiology , RNA Interference , Actins/deficiency , Animals , Gene Knockdown Techniques , Insect Vectors/genetics , Insect Vectors/physiology , Mitochondrial Proton-Translocating ATPases/deficiency
6.
J Clin Invest ; 128(9): 4098-4114, 2018 08 31.
Article En | MEDLINE | ID: mdl-30124467

The host immune system plays a pivotal role in the emergence of tumor cells that are refractory to multiple clinical interventions including immunotherapy, chemotherapy, and radiotherapy. Here, we examined the molecular mechanisms by which the immune system triggers cross-resistance to these interventions. By examining the biological changes in murine and tumor cells subjected to sequential rounds of in vitro or in vivo immune selection via cognate cytotoxic T lymphocytes, we found that multimodality resistance arises through a core metabolic reprogramming pathway instigated by epigenetic loss of the ATP synthase subunit ATP5H, which leads to ROS accumulation and HIF-1α stabilization under normoxia. Furthermore, this pathway confers to tumor cells a stem-like and invasive phenotype. In vivo delivery of antioxidants reverses these phenotypic changes and resensitizes tumor cells to therapy. ATP5H loss in the tumor is strongly linked to failure of therapy, disease progression, and poor survival in patients with cancer. Collectively, our results reveal a mechanism underlying immune-driven multimodality resistance to cancer therapy and demonstrate that rational targeting of mitochondrial metabolic reprogramming in tumor cells may overcome this resistance. We believe these results hold important implications for the clinical management of cancer.


Mitochondria/metabolism , Mitochondrial ADP, ATP Translocases/deficiency , Mitochondrial Proton-Translocating ATPases/deficiency , Neoplasms/metabolism , Neoplasms/therapy , Animals , Antioxidants/administration & dosage , Cell Line, Tumor , Combined Modality Therapy , Drug Resistance, Neoplasm , Epigenesis, Genetic , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mitochondrial ADP, ATP Translocases/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Neoplasms/genetics , Radiation Tolerance , Tumor Escape
7.
Hum Mutat ; 39(4): 563-578, 2018 04.
Article En | MEDLINE | ID: mdl-29314548

In recent years, an increasing number of mitochondrial disorders have been associated with mutations in mitochondrial aminoacyl-tRNA synthetases (mt-aaRSs), which are key enzymes of mitochondrial protein synthesis. Bi-allelic functional variants in VARS2, encoding the mitochondrial valyl tRNA-synthetase, were first reported in a patient with psychomotor delay and epilepsia partialis continua associated with an oxidative phosphorylation (OXPHOS) Complex I defect, before being described in a patient with a neonatal form of encephalocardiomyopathy. Here we provide a detailed genetic, clinical, and biochemical description of 13 patients, from nine unrelated families, harboring VARS2 mutations. All patients except one, who manifested with a less severe disease course, presented at birth exhibiting severe encephalomyopathy and cardiomyopathy. Features included hypotonia, psychomotor delay, seizures, feeding difficulty, abnormal cranial MRI, and elevated lactate. The biochemical phenotype comprised a combined Complex I and Complex IV OXPHOS defect in muscle, with patient fibroblasts displaying normal OXPHOS activity. Homology modeling supported the pathogenicity of VARS2 missense variants. The detailed description of this cohort further delineates our understanding of the clinical presentation associated with pathogenic VARS2 variants and we recommend that this gene should be considered in early-onset mitochondrial encephalomyopathies or encephalocardiomyopathies.


HLA Antigens/genetics , Mitochondrial Encephalomyopathies , Mitochondrial Proton-Translocating ATPases/deficiency , Valine-tRNA Ligase/genetics , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Male , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Encephalomyopathies/physiopathology , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Mutation, Missense , Oxidative Phosphorylation , Phylogeny
8.
Eur J Med Genet ; 60(6): 345-351, 2017 Jun.
Article En | MEDLINE | ID: mdl-28412374

We describe a novel frameshift mutation in the mitochondrial ATP6 gene in a 4-year-old girl associated with ataxia, microcephaly, developmental delay and intellectual disability. A heteroplasmic frameshift mutation in the MT-ATP6 gene was confirmed in the patient's skeletal muscle and blood. The mutation was not detectable in the mother's DNA extracted from blood or buccal cells. Enzymatic and oxymetric analysis of the mitochondrial respiratory system in the patients' skeletal muscle and skin fibroblasts demonstrated an isolated complex V deficiency. Native PAGE with subsequent immunoblotting for complex V revealed impaired complex V assembly and accumulation of ATPase subcomplexes. Whilst northern blotting confirmed equal presence of ATP8/6 mRNA, metabolic 35S-labelling of mitochondrial translation products showed a severe depletion of the ATP6 protein together with aberrant translation product accumulation. In conclusion, this novel isolated complex V defect expands the clinical and genetic spectrum of mitochondrial defects of complex V deficiency. Furthermore, this work confirms the benefit of native PAGE as an additional diagnostic method for the identification of OXPHOS defects, as the presence of complex V subcomplexes is associated with pathogenic mutations of mtDNA.


Ataxia/genetics , Frameshift Mutation , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Ataxia/diagnosis , Cells, Cultured , Child , Female , Fibroblasts/metabolism , Humans , Mitochondrial Encephalomyopathies/diagnosis , Mitochondrial Proton-Translocating ATPases/deficiency , Muscle, Skeletal/metabolism , Syndrome
10.
Biomed Res Int ; 2015: 462592, 2015.
Article En | MEDLINE | ID: mdl-26550569

TMEM70 is involved in the biogenesis of mitochondrial ATP synthase and mutations in the TMEM70 gene impair oxidative phosphorylation. Herein, we report on pathology and treatment of ATP synthase deficiency in four siblings. A consanguineous family of Roma (Gipsy) ethnic origin gave birth to 6 children of which 4 were affected presenting with dysmorphic features, failure to thrive, cardiomyopathy, metabolic crises, and 3-methylglutaconic aciduria as clinical symptoms. Genetic testing revealed a homozygous mutation (c.317-2A>G) in the TMEM70 gene. While light microscopy was unremarkable, ultrastructural investigation of muscle tissue revealed accumulation of swollen degenerated mitochondria with lipid crystalloid inclusions, cristae aggregation, and exocytosis of mitochondrial material. Biochemical analysis of mitochondrial complexes showed an almost complete ATP synthase deficiency. Despite harbouring the same mutation, the clinical outcome in the four siblings was different. Two children died within 60 h after birth; the other two had recurrent life-threatening metabolic crises but were successfully managed with supplementation of anaplerotic amino acids, lipids, and symptomatic treatment during metabolic crisis. In summary, TMEM70 mutations can cause distinct ultrastructural mitochondrial degeneration and almost complete deficiency of ATP synthase but are still amenable to treatment.


Membrane Proteins/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proton-Translocating ATPases/deficiency , Polymorphism, Single Nucleotide/genetics , Adolescent , Child , Diagnosis, Differential , Female , Humans , Male , Mitochondria/enzymology , Mitochondria/ultrastructure , Mitochondrial Diseases/therapy , Mitochondrial Proton-Translocating ATPases/genetics , Treatment Outcome
11.
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
12.
Nat Commun ; 5: 5585, 2014 Dec 18.
Article En | MEDLINE | ID: mdl-25519239

Mitochondrial diseases are systemic, prevalent and often fatal; yet treatments remain scarce. Identifying molecular intervention points that can be therapeutically targeted remains a major challenge, which we confronted via a screening assay we developed. Using yeast models of mitochondrial ATP synthase disorders, we screened a drug repurposing library, and applied genomic and biochemical techniques to identify pathways of interest. Here we demonstrate that modulating the sorting of nuclear-encoded proteins into mitochondria, mediated by the TIM23 complex, proves therapeutic in both yeast and patient-derived cells exhibiting ATP synthase deficiency. Targeting TIM23-dependent protein sorting improves an array of phenotypes associated with ATP synthase disorders, including biogenesis and activity of the oxidative phosphorylation machinery. Our study establishes mitochondrial protein sorting as an intervention point for ATP synthase disorders, and because of the central role of this pathway in mitochondrial biogenesis, it holds broad value for the treatment of mitochondrial diseases.


Membrane Transport Proteins/metabolism , Mitochondrial Diseases/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Nuclear Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Antifungal Agents/pharmacology , Cell Nucleus/metabolism , Databases, Pharmaceutical , Drug Repositioning , Gene Expression Regulation , Humans , Membrane Transport Proteins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Precursor Protein Import Complex Proteins , Mitochondrial Proton-Translocating ATPases/deficiency , Molecular Targeted Therapy , Mutation , Nuclear Proteins/genetics , Oxidative Phosphorylation/drug effects , Protein Transport/drug effects , Pyridines/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction , Thiones/pharmacology
13.
Hum Mol Genet ; 23(10): 2580-92, 2014 May 15.
Article En | MEDLINE | ID: mdl-24399447

Defects of the oxidative phosphorylation system, in particular of cytochrome-c oxidase (COX, respiratory chain complex IV), are common causes of Leigh syndrome (LS), which is a rare neurodegenerative disorder with severe progressive neurological symptoms that usually present during infancy or early childhood. The COX-deficient form of LS is commonly caused by mutations in genes encoding COX assembly factors, e.g. SURF1, SCO1, SCO2 or COX10. However, other mutations affecting genes that encode proteins not directly involved in COX assembly can also cause LS. The leucine-rich pentatricopeptide repeat containing protein (LRPPRC) regulates mRNA stability, polyadenylation and coordinates mitochondrial translation. In humans, mutations in Lrpprc cause the French Canadian type of LS. Despite the finding that LRPPRC deficiency affects the stability of most mitochondrial mRNAs, its pathophysiological effect has mainly been attributed to COX deficiency. Surprisingly, we show here that the impaired mitochondrial respiration and reduced ATP production observed in Lrpprc conditional knockout mouse hearts is caused by an ATP synthase deficiency. Furthermore, the appearance of inactive subassembled ATP synthase complexes causes hyperpolarization and increases mitochondrial reactive oxygen species production. Our findings shed important new light on the bioenergetic consequences of the loss of LRPPRC in cardiac mitochondria.


Mitochondria, Heart/enzymology , Mitochondrial Proton-Translocating ATPases/deficiency , Neoplasm Proteins/genetics , Adenosine Triphosphate/biosynthesis , Animals , Electron Transport Complex IV/metabolism , Energy Metabolism , Humans , Leigh Disease/genetics , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/pathology , Mitochondrial Proton-Translocating ATPases/genetics , Oxidative Phosphorylation , Oxygen Consumption , Protein Multimerization , Reactive Oxygen Species/metabolism
14.
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
15.
Neurogenetics ; 13(4): 375-86, 2012 Nov.
Article En | MEDLINE | ID: mdl-22986587

Mammalian complex V (F1F0-ATP synthase or ATPase) uses the proton gradient to generate ATP during oxidative phosphorylation and requires several helper proteins, including TMEM70, to form the holoenzyme in a stepwise process in which nuclear DNA is combined with mitochondrial DNA-encoded subunits. We report the clinical and molecular findings in three patients presenting lactic acidosis, 3-methylglutaconic aciduria, and hypertrophic cardiomyopathy. All three showed an isolated defect of fully assembled ATP synthase in association with a "common" (c.317-2A > G) and a new (c.628A > C/p.T210P) variant in TMEM70. Interestingly, one of the patients also showed nitric oxide-responsive pulmonary arterial hypertension, a finding never before associated with TMEM70 deficiency. In addition to widening the clinical and mutational spectrum of defective ATP synthase, our study also suggests that mutant TMEM70 associates in high molecular weight complexes (470-550 kDa) when expressed in Hela cells and exerts a direct action in ATP synthase biogenesis and assembly, mediating the incorporation of F1 moieties.


Adenosine Triphosphatases/biosynthesis , Carrier Proteins/biosynthesis , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proton-Translocating ATPases/deficiency , Mutation , Acidosis, Lactic/genetics , Acidosis, Lactic/metabolism , Base Sequence , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Cerebellar Ataxia/genetics , Child , Child, Preschool , Familial Primary Pulmonary Hypertension , Female , Fibroblasts/metabolism , HeLa Cells , Humans , Hypertension, Pulmonary/genetics , Infant , Metabolism, Inborn Errors/genetics , Molecular Sequence Data , Sequence Analysis, DNA , Skin/metabolism
17.
Biochim Biophys Acta ; 1817(7): 1037-43, 2012 Jul.
Article En | MEDLINE | ID: mdl-22433607

Early onset mitochondrial encephalo-cardiomyopathy due to isolated deficiency of ATP synthase is frequently caused by mutations in TMEM70 gene encoding enzyme-specific ancillary factor. Diminished ATP synthase results in low ATP production, elevated mitochondrial membrane potential and increased ROS production. To test whether the patient cells may react to metabolic disbalance by changes in oxidative phosphorylation system, we performed a quantitative analysis of respiratory chain complexes and intramitochondrial proteases involved in their turnover. SDS- and BN-PAGE Western blot analysis of fibroblasts from 10 patients with TMEM70 317-2A>G homozygous mutation showed a significant 82-89% decrease of ATP synthase and 50-162% increase of respiratory chain complex IV and 22-53% increase of complex III. The content of Lon protease, paraplegin and prohibitins 1 and 2 was not significantly changed. Whole genome expression profiling revealed a generalized upregulation of transcriptional activity, but did not show any consistent changes in mRNA levels of structural subunits, specific assembly factors of respiratory chain complexes, or in regulatory genes of mitochondrial biogenesis which would parallel the protein data. The mtDNA content in patient cells was also not changed. The results indicate involvement of posttranscriptional events in the adaptive regulation of mitochondrial biogenesis that allows for the compensatory increase of respiratory chain complexes III and IV in response to deficiency of ATP synthase.


Electron Transport Complex III/metabolism , Electron Transport Complex IV/metabolism , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proton-Translocating ATPases/deficiency , Mutation/genetics , Up-Regulation , DNA, Mitochondrial/metabolism , Electron Transport/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Humans , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Oxidative Phosphorylation , Peptide Hydrolases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
Mitochondrion ; 12(1): 149-55, 2012 Jan.
Article En | MEDLINE | ID: mdl-21742063

Mitochondrial chronic stress that originates from defective mitochondria is implicated in a growing list of human diseases. To enhance understanding of pathophysiology of chronic mitochondrial dysfunction we investigated human osteosarcoma cells with 2 types of chronic stress: corresponding to the mutation in ATP synthase subunit 6 encoded by mtDNA (NARP syndrome-mild stress) and to a total lack of mtDNA (Rho0 cells-heavy stress). We previously found that selenium influenced mitochondrial stress response and lowered ROS production. Therefore, in this study effect of selenite on other mitochondrial parameters was investigated. We showed that presence of selenium improved survival of starved cells, modified organization of mitochondrial network in NARP cybrids and decreased cytosolic calcium level in NARP and Rho0 cells. Selenium did not affect mitochondrial membrane potential, ATP level, activity of ATP synthase and activity of complex II of the respiratory chain.


Mitochondria/drug effects , Mitochondria/physiology , Sodium Selenite/metabolism , Adenosine Triphosphate/analysis , Calcium/analysis , Cell Line, Tumor , Cell Respiration/drug effects , Cell Survival/drug effects , Cytosol/chemistry , DNA, Mitochondrial/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/enzymology , Mitochondria/genetics , Mitochondrial Myopathies/physiopathology , Mitochondrial Proton-Translocating ATPases/deficiency , Retinitis Pigmentosa/physiopathology
19.
J Cell Biochem ; 113(1): 141-7, 2012 Jan.
Article En | MEDLINE | ID: mdl-21882222

Proton pump inhibitors (PPIs) are widely used against gastroesophageal reflux disease. Recent epidemiological studies suggest that PPI users have an increased risk of fractures, but a causal relationship has been questioned. We have therefore investigated the skeletal phenotype in H(+) /K(+) ATPase beta-subunit knockout (KO) female mice. Skeletal parameters were determined in 6- and 20-month-old KO mice and in wild-type controls (WT). Whole body bone mineral density (BMD) and bone mineral content (BMC) were measured by dual energy X-ray absorptiometry (DXA). Femurs were examined with µCT analyses and break force were examined by a three-point bending test. Plasma levels of gastrin, RANKL, OPG, osteocalcin, leptin, and PTH were analyzed. KO mice had lower whole body BMC at 6 months (0.53 vs. 0.59 g, P = 0.035) and at 20 months (0.49 vs. 0.74 g, P < 0.01) compared to WT as well as lower BMD at 6 months (0.068 vs. 0.072 g/cm(2) , P = 0.026) and 20 months (0.067 vs. 0.077 g/cm(2) , P < 0.01). Mechanical strength was lower in KO mice at the age of 20 months (6.7 vs. 17.9 N, P < 0.01). Cortical thickness at 20 months and trabecular bone volume% at 6 months were significantly reduced in KO mice. Plasma OPG/RANKL ratio and PTH was increased in KO mice compared to controls. H(+) /K(+) ATPase beta subunit KO mice had decreased BMC and BMD, reduced cortical thickness and inferior mechanical bone strength. Whereas the mechanism is uncertain, these findings suggest a causal relationship between long-term PPI use and an increased risk of fractures.


Bone Density/genetics , Bone and Bones/pathology , Mitochondrial Proton-Translocating ATPases/deficiency , Mitochondrial Proton-Translocating ATPases/genetics , Absorptiometry, Photon/methods , Animals , Female , Fractures, Bone/chemically induced , Gastrins/blood , Leptin/blood , Mice , Mice, Inbred BALB C , Mice, Knockout , Osteocalcin/blood , Osteoprotegerin/blood , Parathyroid Hormone/blood , Protein Subunits/deficiency , Protein Subunits/genetics , Proton Pump Inhibitors/adverse effects , Proton Pump Inhibitors/therapeutic use , RANK Ligand/blood , Risk
...