Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
BMC Infect Dis ; 22(1): 188, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35209862

ABSTRACT

BACKGROUND: Mitochondrial myopathy caused by the long-term use of nucleos(t)ide analogue in patients with chronic hepatitis B (CHB) is mostly characterized by myasthenia and myalgia. Cases with respiratory failure as the prominent manifestation and multisystem symptoms have not been reported. CASE REPORT: We report a case of mitochondrial myopathy associated with the long-term use of entecavir for CHB. The patient was a 54-year-old male who was treated with entecavir for 9 years. During the treatment, hepatitis B virus (HBV) DNA was negative and liver function was normal. However, generalized fatigue, poor appetite, dysosmia and other discomforts gradually presented starting at the 5th year of treatment, and respiratory failure was the prominent manifestation in the later stage of disease progression. The diagnosis was based on histopathology examination. The dysosmia, hypoxemia and digestive tract symptoms were gradually improved after withdrawal of entecavir. DISCUSSION: Mitochondrial myopathy is a rare side effect of entecavir and can be diagnosed by muscle biopsy. Although the incidence is extremely low, but the severe cases can lead to respiratory failure. We should receive adequate attention in clinical practice.


Subject(s)
Hepatitis B, Chronic , Mitochondrial Myopathies , Respiratory Insufficiency , Antiviral Agents/adverse effects , Guanine/analogs & derivatives , Hepatitis B virus/genetics , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/drug therapy , Humans , Male , Middle Aged , Mitochondrial Myopathies/chemically induced , Mitochondrial Myopathies/complications , Mitochondrial Myopathies/drug therapy , Respiratory Insufficiency/drug therapy , Treatment Outcome
2.
J Inherit Metab Dis ; 44(5): 1186-1198, 2021 09.
Article in English | MEDLINE | ID: mdl-33934389

ABSTRACT

Mitochondrial myopathies (MM) are caused by mutations that typically affect genes involved in oxidative phosphorylation. Main symptoms are exercise intolerance and fatigue. Currently, there is no specific treatment for MM. Resveratrol (RSV) is a nutritional supplement that in preclinical studies has been shown to stimulate mitochondrial function. We hypothesized that RSV could improve exercise capacity in patients with MM. The study design was randomized, double-blind, cross-over and placebo-controlled. Eleven patients with genetically verified MM were randomized to receive either 1000 mg/day RSV or placebo (P) for 8 weeks followed by a 4-week washout and then the opposite treatment. Primary outcomes were changes in heart rate (HR) during submaximal cycling exercise and peak oxygen utilization (VO2 max) during maximal exercise. Secondary outcomes included reduction in perceived exertion, changes in lactate concentrations, self-rated function (SF-36) and fatigue scores (FSS), activities of electron transport chain complexes I and IV in mononuclear cells and mitochondrial biomarkers in muscle tissue among others. There were no significant differences in primary and secondary outcomes between treatments. Mean HR changes were -0.3 ± 4.3 (RSV) vs 1.8 ± 5.0 bpm (P), P = .241. Mean VO2 max changes were 0.7 ± 1.4 (RSV) vs -0.2 ± 2.3 mL/min/kg (P), P = .203. The study provides evidence that 1000 mg RSV daily is ineffective in improving exercise capacity in adults with MM. These findings indicate that previous in vitro studies suggesting a therapeutic potential for RSV in MM, do not translate into clinically meaningful effects in vivo.


Subject(s)
Mitochondrial Myopathies/drug therapy , Resveratrol/therapeutic use , Adult , Aged , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Resveratrol/pharmacology
3.
Curr Opin Neurol ; 32(5): 715-721, 2019 10.
Article in English | MEDLINE | ID: mdl-31408013

ABSTRACT

PURPOSE OF REVIEW: Although mitochondrial diseases impose a significant functional limitation in the lives of patients, treatment of these conditions has been limited to dietary supplements, exercise, and physical therapy. In the past few years, however, translational medicine has identified potential therapies for these patients. RECENT FINDINGS: For patients with primary mitochondrial myopathies, preliminary phase I and II multicenter clinical trials of elamipretide indicate safety and suggest improvement in 6-min walk test (6MWT) performance and fatigue scales. In addition, for thymidine kinase 2-deficient (TK2d) myopathy, compassionate-use oral administration of pyrimidine deoxynucleosides have shown preliminary evidence of safety and efficacy in survival of early onset patients and motor functions relative to historical TK2d controls. SUMMARY: The prospects of effective therapies that improve the quality of life for patients with mitochondrial myopathy underscore the necessity for definitive diagnoses natural history studies for better understanding of the diseases.


Subject(s)
Mitochondrial Myopathies/drug therapy , Oligopeptides/therapeutic use , Quality of Life , Clinical Trials as Topic , Exercise/physiology , Fatigue/physiopathology , Humans , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/physiopathology
4.
Hum Mol Genet ; 25(15): 3178-3191, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288451

ABSTRACT

Acute pharmacological activation of adenosine monophosphate (AMP)-kinase using 5-aminoimidazole-4-carboxamide-1-b-D-ribofuranoside (AICAR) has been shown to improve muscle mitochondrial function by increasing mitochondrial biogenesis. We asked whether prolonged AICAR treatment is beneficial in a mouse model of slowly progressing mitochondrial myopathy (Cox10-Mef2c-Cre), and whether the compensatory mechanism is indeed an increase in mitochondrial biogenesis. We treated the animals for 3 months and found that sustained AMP-dependent kinase activation improved cytochrome c oxidase activity, rescued the motor phenotype and delayed the onset of the myopathy. This improvement was observed whether treatment started before or after the onset of the disease. We found that AICAR increased skeletal muscle regeneration thereby decreasing the levels of deleted Cox10-floxed alleles. We conclude that although increase in mitochondrial biogenesis and other pathways may contribute, the main mechanism by which AICAR improves the myopathy phenotype is by promoting muscle regeneration.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Mitochondrial Myopathies/drug therapy , Mitochondrial Myopathies/metabolism , Muscle Fibers, Skeletal/physiology , Regeneration/drug effects , Ribonucleotides/pharmacology , AMP-Activated Protein Kinases/genetics , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Disease Models, Animal , Enzyme Activation/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/pathology
5.
Pharmacol Res ; 138: 37-42, 2018 12.
Article in English | MEDLINE | ID: mdl-30267763

ABSTRACT

Mitochondrial myopathy (MM) is characterised by muscle weakness, exercise intolerance and various histopathological changes. Recently, a subset of MM has also been associated with aberrant activation of mammalian target of rapamycin complex 1 (mTORC1) in skeletal muscle. This aberrant mTORC1 activation promotes increased de novo nucleotide synthesis, which contributes to abnormal expansion and imbalance of skeletal muscle deoxyribonucleoside triphosphates (dNTP) pools. However, the exact mechanism via which mTORC1-stimulated de novo nucleotide biosynthesis ultimately disturbs muscle dNTP pools remains unclear. In this article, it is proposed that mTORC1-stimulated de novo nucleotide synthesis in skeletal muscle cells with respiratory chain dysfunction promotes an asymmetric increase of purine nucleotides, probably due to NAD+ deficiency. This in turn could disrupt purine nucleotide-dependent allosteric feedback regulatory mechanisms, ultimately leading to dNTP pools aberration. Pharmacological down-modulation of aminoimidazole carboxamide ribonucleotide transformylase/inosine monophosphate cyclohydrolase (ATIC) activity is also proposed as a potential therapeutic strategy in MM exhibiting mTORC1-driven abnormal metabolic reprogramming, including aberrant dNTPs pools.


Subject(s)
Mitochondrial Myopathies/metabolism , Purine Nucleotides/metabolism , Animals , Humans , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Hydroxymethyl and Formyl Transferases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitochondrial Myopathies/drug therapy , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Nucleotide Deaminases/antagonists & inhibitors , Nucleotide Deaminases/metabolism
6.
Hum Mol Genet ; 24(23): 6580-7, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26374844

ABSTRACT

Muscle weakness and exercise intolerance are hallmark symptoms in mitochondrial disorders. Little is known about the mechanisms leading to impaired skeletal muscle function and ultimately muscle weakness in these patients. In a mouse model of lethal mitochondrial myopathy, the muscle-specific Tfam knock-out (KO) mouse, we previously demonstrated an excessive mitochondrial Ca(2+) uptake in isolated muscle fibers that could be inhibited by the cyclophilin D (CypD) inhibitor, cyclosporine A (CsA). Here we show that the Tfam KO mice have increased CypD levels, and we demonstrate that this increase is a common feature in patients with mitochondrial myopathy. We tested the effect of CsA treatment on Tfam KO mice during the transition from a mild to terminal myopathy. CsA treatment counteracted the development of muscle weakness and improved muscle fiber Ca(2+) handling. Importantly, CsA treatment prolonged the lifespan of these muscle-specific Tfam KO mice. These results demonstrate that CsA treatment is an efficient therapeutic strategy to slow the development of severe mitochondrial myopathy.


Subject(s)
Cyclophilins/antagonists & inhibitors , Cyclosporine/therapeutic use , Mitochondria/metabolism , Mitochondrial Myopathies/drug therapy , Muscle, Skeletal/metabolism , Animals , Calcium/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/drug effects , Cyclophilins/genetics , DNA, Mitochondrial , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/metabolism , Muscle, Skeletal/drug effects , Mutation
7.
Am J Physiol Regul Integr Comp Physiol ; 312(5): R689-R701, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28179228

ABSTRACT

Muscle weakness and exercise intolerance negatively affect the quality of life of patients with mitochondrial myopathy. Short-term dietary nitrate supplementation has been shown to improve exercise performance and reduce oxygen cost of exercise in healthy humans and trained athletes. We investigated whether 1 wk of dietary inorganic nitrate supplementation decreases the oxygen cost of exercise and improves mitochondrial function in patients with mitochondrial myopathy. Ten patients with mitochondrial myopathy (40 ± 5 yr, maximal whole body oxygen uptake = 21.2 ± 3.2 ml·min-1·kg body wt-1, maximal work load = 122 ± 26 W) received 8.5 mg·kg body wt-1·day-1 inorganic nitrate (~7 mmol) for 8 days. Whole body oxygen consumption at 50% of the maximal work load, in vivo skeletal muscle oxidative capacity (evaluated from postexercise phosphocreatine recovery using 31P-magnetic resonance spectroscopy), and ex vivo mitochondrial oxidative capacity in permeabilized skinned muscle fibers (measured with high-resolution respirometry) were determined before and after nitrate supplementation. Despite a sixfold increase in plasma nitrate levels, nitrate supplementation did not affect whole body oxygen cost during submaximal exercise. Additionally, no beneficial effects of nitrate were found on in vivo or ex vivo muscle mitochondrial oxidative capacity. This is the first time that the therapeutic potential of dietary nitrate for patients with mitochondrial myopathy was evaluated. We conclude that 1 wk of dietary nitrate supplementation does not reduce oxygen cost of exercise or improve mitochondrial function in the group of patients tested.


Subject(s)
Exercise , Mitochondria, Muscle/metabolism , Mitochondrial Myopathies/drug therapy , Mitochondrial Myopathies/physiopathology , Nitrates/administration & dosage , Oxygen Consumption/drug effects , Administration, Oral , Adult , Aged , Exercise Tolerance/drug effects , Female , Humans , Male , Middle Aged , Mitochondria, Muscle/drug effects , Muscle Strength/drug effects , Psychomotor Performance/drug effects , Treatment Outcome , Young Adult
8.
J Inherit Metab Dis ; 40(6): 831-843, 2017 11.
Article in English | MEDLINE | ID: mdl-28871440

ABSTRACT

BACKGROUND: Observational reports suggest that supplementation that increases citric acid cycle intermediates via anaplerosis may have therapeutic advantages over traditional medium-chain triglyceride (MCT) treatment of long-chain fatty acid oxidation disorders (LC-FAODs) but controlled trials have not been reported. The goal of our study was to compare the effects of triheptanoin (C7), an anaplerotic seven-carbon fatty acid triglyceride, to trioctanoin (C8), an eight-carbon fatty acid triglyceride, in patients with LC-FAODs. METHODS: A double blinded, randomized controlled trial of 32 subjects with LC-FAODs (carnitine palmitoyltransferase-2, very long-chain acylCoA dehydrogenase, trifunctional protein or long-chain 3-hydroxy acylCoA dehydrogenase deficiencies) who were randomly assigned a diet containing 20% of their total daily energy from either C7 or C8 for 4 months was conducted. Primary outcomes included changes in total energy expenditure (TEE), cardiac function by echocardiogram, exercise tolerance, and phosphocreatine recovery following acute exercise. Secondary outcomes included body composition, blood biomarkers, and adverse events, including incidence of rhabdomyolysis. RESULTS: Patients in the C7 group increased left ventricular (LV) ejection fraction by 7.4% (p = 0.046) while experiencing a 20% (p = 0.041) decrease in LV wall mass on their resting echocardiogram. They also required a lower heart rate for the same amount of work during a moderate-intensity exercise stress test when compared to patients taking C8. There was no difference in TEE, phosphocreatine recovery, body composition, incidence of rhabdomyolysis, or any secondary outcome measures between the groups. CONCLUSIONS: C7 improved LV ejection fraction and reduced LV mass at rest, as well as lowering heart rate during exercise among patients with LC-FAODs. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov NCT01379625.


Subject(s)
Caprylates/therapeutic use , Cardiomyopathies/drug therapy , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/drug therapy , Mitochondrial Myopathies/drug therapy , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/drug therapy , Rhabdomyolysis/drug therapy , Triglycerides/therapeutic use , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Adolescent , Adult , Cardiomyopathies/metabolism , Carnitine/metabolism , Child , Dietary Fats/metabolism , Double-Blind Method , Exercise/physiology , Female , Humans , Lipid Metabolism, Inborn Errors/metabolism , Male , Middle Aged , Mitochondrial Myopathies/metabolism , Mitochondrial Trifunctional Protein/metabolism , Nervous System Diseases/metabolism , Oxidation-Reduction , Rhabdomyolysis/metabolism , Young Adult
9.
Eur J Neurol ; 24(4): 587-593, 2017 04.
Article in English | MEDLINE | ID: mdl-28181352

ABSTRACT

BACKGROUND AND PURPOSE: Most mitochondrial disorders with onset in early childhood are progressive and involve multiple organs. The m.3250T>C mutation in MTTL1 has previously been described in a few individuals with a possibly riboflavin-responsive myopathy and an association with sudden infant death syndrome was suspected. We describe a large family with this mutation and evaluate the effect of riboflavin treatment. METHODS: Medical data were collected with the help of a standardized data collection form. Sanger sequencing was used to screen for variants in mitochondrial DNA and the proportion of the mutation was analyzed in different tissues. Biochemical and muscle morphological investigations of muscle tissue were performed in two individuals. The effect of riboflavin treatment was evaluated in two individuals. RESULTS: Thirteen family members experienced exercise intolerance with fatigue and weakness. Inheritance was maternal with 100% penetrance. The course was either static or showed improvement over time. There was no evidence of other organ involvement except for a possible mild transient cardiac enlargement in one child. Muscle investigations showed isolated complex I deficiency and mitochondrial proliferation. The level of m.3250T>C was apparently 100%, i.e. homoplasmic, in all examined tissues. Riboflavin treatment showed no effect in any treated family member and there have been no cases of sudden infant death in this family. CONCLUSIONS: This study illustrates the importance of considering mitochondrial disorders in the work-up of individuals with exercise intolerance and provides a better understanding of the phenotype associated with the m.3250T>C mutation in MTTL1.


Subject(s)
DNA, Mitochondrial/genetics , Exercise Tolerance/genetics , Mitochondrial Myopathies/genetics , Mutation , RNA, Transfer/genetics , Adult , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Mitochondria/genetics , Mitochondrial Myopathies/drug therapy , Pedigree , Phenotype , Riboflavin/therapeutic use , Vitamin B Complex/therapeutic use , Young Adult
10.
J Inherit Metab Dis ; 39(1): 47-58, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26109258

ABSTRACT

Mitochondrial trifunctional protein (MTP) deficiency caused by HADHA or HADHB gene mutations exhibits substantial molecular, biochemical, and clinical heterogeneity and ranks among the more severe fatty acid oxidation (FAO) disorders, without pharmacological treatment. Since bezafibrate has been shown to potentially correct other FAO disorders in patient cells, we analyzed its effects in 26 MTP-deficient patient fibroblasts representing 16 genotypes. Overall, the patient cell lines exhibited variable, complex, biochemical profiles and pharmacological responses. HADHA-deficient fibroblasts showed markedly reduced alpha subunit protein levels together with decreased beta-subunit abundance, exhibited a -86 to -96% defect in LCHAD activity, and produced large amounts of C14 and C16 hydroxyacylcarnitines. In control fibroblasts, exposure to bezafibrate (400 µM for 48 h) increased the abundance of HADHA and HADHB mRNAs, immune-detectable alpha and beta subunit proteins, activities of LCHAD and LCKAT, and stimulated FAO capacities, clearly indicating that MTP is pharmacologically up-regulated by bezafibrate in human fibroblasts. In MTP-deficient patient fibroblasts, which were found markedly FAO-deficient, bezafibrate improved FAO capacities in six of 26 (23%) cases, including three cell lines heterozygous for the common c1528G > C mutation. Altogether, our results strongly suggest that, due to variable effects of HADHA and HADHB mutations on MTP abundance and residual activity, improvement of MTP deficiency in response to bezafibrate was achieved in a subset of responsive genotypes.


Subject(s)
Bezafibrate/pharmacology , Cardiomyopathies/drug therapy , Fibroblasts/drug effects , Hypolipidemic Agents/pharmacology , Lipid Metabolism, Inborn Errors/drug therapy , Mitochondrial Myopathies/drug therapy , Mitochondrial Trifunctional Protein, alpha Subunit/deficiency , Mitochondrial Trifunctional Protein, beta Subunit/deficiency , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/drug therapy , Rhabdomyolysis/drug therapy , Cardiomyopathies/genetics , Cell Line , Genotype , Humans , Lipid Metabolism, Inborn Errors/genetics , Mitochondrial Myopathies/genetics , Mitochondrial Trifunctional Protein/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, beta Subunit/genetics , Mutation/genetics , Nervous System Diseases/genetics , Rhabdomyolysis/genetics
11.
Hum Mol Genet ; 21(3): 526-35, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22012983

ABSTRACT

Mitochondrial dysfunction is an important cause of metabolic disorders of children and adults, with no effective therapy options. Recently, induction of mitochondrial biogenesis, by transgenic overexpression of PGC1-alpha [peroxisome proliferator-activated receptor (PPAR)-gamma coactivator 1-alpha], was reported to delay progression of early-onset cytochrome-c-oxidase (COX) deficiency in skeletal muscle of two mouse models: a muscle-specific knock-out of COX10 (COX10-mKO) and a constitutive knock-out of Surf1 (Surf1-KO). A pan-PPAR agonist, bezafibrate, could similarly delay myopathy progression in COX10-mKOs, but not in SURF1-KOs. We asked whether bezafibrate affected disease progression in late-onset adult-type mitochondrial myopathy mice. These 'Deletor mice' express a dominant patient mutation in Twinkle-helicase, leading to accumulation of multiple mtDNA deletions and subsequent progressive respiratory chain (RC) deficiency with COX-negative muscle fibers at 12 months of age. The primary and secondary molecular findings in Deletor mice mimic closely those in patients with Twinkle myopathy. We applied 0.5% bezafibrate diet to Deletors for 22 weeks, starting at disease manifestation, mimicking patient treatment after diagnosis. Bezafibrate delayed significantly the accumulation of COX-negative fibers and multiple mtDNA deletions. However, mitochondrial biogenesis was not induced: mitochondrial DNA copy number, transcript and RC protein amounts decreased in both Deletors and wild-type mice. Furthermore, bezafibrate induced severe lipid oxidation effects, with hepatomegaly and loss of adipose tissue, the mechanism involving lipid mobilization by high hepatic expression of FGF21 cytokine. However, as bezafibrate has been tolerated well by humans, the beneficial muscle findings in Deletor mice support consideration of bezafibrate trials on adult patients with mitochondrial myopathy.


Subject(s)
Bezafibrate/therapeutic use , Mitochondrial Myopathies/drug therapy , Adipose Tissue/drug effects , Animals , Body Temperature/drug effects , DNA Helicases/genetics , DNA, Mitochondrial/analysis , Disease Progression , Electron Transport Complex IV/analysis , Female , Fibroblast Growth Factors/metabolism , Hepatomegaly/chemically induced , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mitochondria/metabolism , Mitochondrial Myopathies/enzymology , Mitochondrial Myopathies/metabolism , Mitochondrial Proteins/genetics , Muscle Fibers, Skeletal/enzymology , Mutation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Peroxisome Proliferator-Activated Receptors/agonists , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Messenger/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Weight Loss
12.
Proc Natl Acad Sci U S A ; 108(29): 11989-94, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21715656

ABSTRACT

Due to the lack of relevant animal models, development of effective treatments for human mitochondrial diseases has been limited. Here we establish a rapid, yeast-based assay to screen for drugs active against human inherited mitochondrial diseases affecting ATP synthase, in particular NARP (neuropathy, ataxia, and retinitis pigmentosa) syndrome. This method is based on the conservation of mitochondrial function from yeast to human, on the unique ability of yeast to survive without production of ATP by oxidative phosphorylation, and on the amenability of the yeast mitochondrial genome to site-directed mutagenesis. Our method identifies chlorhexidine by screening a chemical library and oleate through a candidate approach. We show that these molecules rescue a number of phenotypes resulting from mutations affecting ATP synthase in yeast. These compounds are also active on human cybrid cells derived from NARP patients. These results validate our method as an effective high-throughput screening approach to identify drugs active in the treatment of human ATP synthase disorders and suggest that this type of method could be applied to other mitochondrial diseases.


Subject(s)
Chlorhexidine/pharmacology , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Mitochondrial Myopathies/drug therapy , Mitochondrial Proton-Translocating ATPases/genetics , Oleic Acid/pharmacology , Retinitis Pigmentosa/drug therapy , Cell Line , Chlorhexidine/therapeutic use , Gene Expression Profiling , Humans , Mutagenesis, Site-Directed , Mutation/genetics , Oleic Acid/therapeutic use , Saccharomycetales
13.
Med Sci (Paris) ; 30(12): 1161-8, 2014 Dec.
Article in French | MEDLINE | ID: mdl-25537047

ABSTRACT

Since its discovery and description by Louis Pasteur, the budding yeast Saccharomyces cerevisiae, which was used for thousands of years for alcoholic fermentation and as a leavening agent, has become a popular model system in biology. One of the reasons for this popularity is the strong conservation from yeast to human of most of the pathways controlling cell growth and fate. In addition, at least 30 % of human genes involved in diseases have a functional homolog in yeast. Hence, yeast is now widely used for modelling and deciphering physiopathological mechanisms as well as for developing pharmacological approaches like phenotype-based drug screening. Three examples of such yeast-based chemobiological studies are presented.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Biological , Saccharomyces cerevisiae , Animals , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/immunology , Humans , Mice , Mitochondrial Diseases/drug therapy , Mitochondrial Myopathies/drug therapy , Phenotype , Prion Diseases/drug therapy , Retinitis Pigmentosa/drug therapy , Saccharomyces cerevisiae/genetics
14.
Neuromuscul Disord ; 43: 14-19, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39173541

ABSTRACT

Myopathy is a common manifestation in mitochondrial disorders, but the pathomechanisms are still insufficiently studied in children. Here, we report a severe, progressive mitochondrial myopathy in a four-year-old child, who died at eight years. He developed progressive loss of muscle strength with nocturnal hypoventilation and dilated cardiomyopathy. Skeletal muscle showed ragged red fibers and severe combined respiratory chain deficiency. Mitochondrial DNA sequencing revealed a novel m.5670A>G mutation in mitochondrial tRNAAsn (MTTN) with 88 % heteroplasmy in muscle. The proband also had systemic NAD+ deficiency but rescuing this with the NAD+ precursor niacin did not stop disease progression. Targeted metabolomics revealed an overall shift of metabolism towards controls after niacin supplementation, with normalized tryptophan metabolites and lipid-metabolic markers, but most amino acids did not respond to niacin therapy. To conclude, we report a new MTTN mutation, secondary NAD+ deficiency in childhood-onset mitochondrial myopathy with metabolic but meager clinical response to niacin supplementation.


Subject(s)
Mitochondrial Myopathies , NAD , Niacin , Humans , Male , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/drug therapy , NAD/metabolism , Child, Preschool , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Mutation , Dietary Supplements , DNA, Mitochondrial/genetics , Child
15.
J Pineal Res ; 54(1): 107-25, 2013 Jan.
Article in English | MEDLINE | ID: mdl-24446866

ABSTRACT

Mitochondrial dysfunction is a hallmark of amyloid ß-peptide (Aß)-induced neurodegeneration of Alzheimer's disease (AD). This study investigated whether mtDNA T8993G mutation-induced complex V inhibition, clinically associated with neurological muscle weakness, ataxia, and retinitis pigmentosa (NARP), is a potential risk factor for AD and the pathological link for long-term exposure of Aß-induced mitochondrial toxicity and apoptosis in NARP cybrids. Using noninvasive fluorescence probe-coupled laser scanning imaging microscopy and NARP cybrids harboring 98% mutant genes along with its parental 143B osteosarcoma cells, we demonstrated that Aß-augmented mitochondrial Ca(2+) (mCa(2+))-independent mitochondrial reactive oxygen species (mROS) formation for a cardiolipin (CL, a major mitochondrial protective phospholipid)-dependent lethal modulation of the mitochondrial permeability transition (MPT). Aß augmented not only the amount but also the propagation rate of mROS-induced mROS formation to significantly depolarize mitochondrial membrane potential (∆Ψ(m)) and reduce mCa(2+) stress. Aß-augmented mROS oxidized and depleted CL, thereby enhances mitochondrial fission and movement retardation, which promoted the NARP-augmented lethal transient-MPT (t-MPT) to switch to its irreversible mode of permanent-MPT (p-MPT). Interestingly, melatonin, a multiple mitochondrial protector, markedly reduced Aß-augmented mROS formation and therefore significantly reduced mROS-mediated depolarization of ∆Ψ(m), fission of mitochondria and retardation of mitochondrial movement to stabilize CL and hence the MPT. In the presence of melatonin, Aß-promoted p-MPT was reversed to a protective t-MPT, which preserved ∆Ψ(m) and lowered elevated mCa(2+) to sublethal levels for an enhanced mCa(2+)-dependent O(2) consumption. Thus, melatonin may potentially rescue AD patients associated with NARP symptoms.


Subject(s)
Amyloid beta-Peptides/pharmacology , Cardiolipins/metabolism , Melatonin/therapeutic use , Mitochondrial Membrane Transport Proteins/drug effects , Calcium/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondrial Myopathies/drug therapy , Mitochondrial Permeability Transition Pore , Reactive Oxygen Species/metabolism , Retinitis Pigmentosa/drug therapy
16.
Neurology ; 101(3): e238-e252, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37268435

ABSTRACT

BACKGROUND AND OBJECTIVES: Primary mitochondrial myopathies (PMMs) encompass a group of genetic disorders that impair mitochondrial oxidative phosphorylation, adversely affecting physical function, exercise capacity, and quality of life (QoL). Current PMM standards of care address symptoms, with limited clinical impact, constituting a significant therapeutic unmet need. We present data from MMPOWER-3, a pivotal, phase-3, randomized, double-blind, placebo-controlled clinical trial that evaluated the efficacy and safety of elamipretide in participants with genetically confirmed PMM. METHODS: After screening, eligible participants were randomized 1:1 to receive either 24 weeks of elamipretide at a dose of 40 mg/d or placebo subcutaneously. Primary efficacy endpoints included change from baseline to week 24 on the distance walked on the 6-minute walk test (6MWT) and total fatigue on the Primary Mitochondrial Myopathy Symptom Assessment (PMMSA). Secondary endpoints included most bothersome symptom score on the PMMSA, NeuroQoL Fatigue Short-Form scores, and the patient global impression and clinician global impression of PMM symptoms. RESULTS: Participants (N = 218) were randomized (n = 109 elamipretide; n = 109 placebo). The m0ean age was 45.6 years (64% women; 94% White). Most of the participants (n = 162 [74%]) had mitochondrial DNA (mtDNA) alteration, with the remainder having nuclear DNA (nDNA) defects. At screening, the most frequent bothersome PMM symptom on the PMMSA was tiredness during activities (28.9%). At baseline, the mean distance walked on the 6MWT was 336.7 ± 81.2 meters, the mean score for total fatigue on the PMMSA was 10.6 ± 2.5, and the mean T score for the Neuro-QoL Fatigue Short-Form was 54.7 ± 7.5. The study did not meet its primary endpoints assessing changes in the 6MWT and PMMSA total fatigue score (TFS). Between the participants receiving elamipretide and those receiving placebo, the difference in the least squares mean (SE) from baseline to week 24 on distance walked on the 6MWT was -3.2 (95% CI -18.7 to 12.3; p = 0.69) meters, and on the PMMSA, the total fatigue score was -0.07 (95% CI -0.10 to 0.26; p = 0.37). Elamipretide treatment was well-tolerated with most adverse events being mild to moderate in severity. DISCUSSION: Subcutaneous elamipretide treatment did not improve outcomes in the 6MWT and PMMSA TFS in patients with PMM. However, this phase-3 study demonstrated that subcutaneous elamipretide is well-tolerated. TRIAL REGISTRATION INFORMATION: Trial registered with clinicaltrials.gov, Clinical Trials Identifier: NCT03323749; submitted on October 12, 2017; first patient enrolled October 9, 2017. CLINICALTRIALS: gov/ct2/show/NCT03323749?term = elamipretide&draw = 2&rank = 9. CLASSIFICATION OF EVIDENCE: This study provides Class I evidence that elamipretide does not improve the 6MWT or fatigue at 24 weeks compared with placebo in patients with primary mitochondrial myopathy.


Subject(s)
Mitochondrial Myopathies , Quality of Life , Humans , Female , Middle Aged , Male , Merozoite Surface Protein 1/therapeutic use , Mitochondrial Myopathies/drug therapy , Fatigue , Double-Blind Method , Treatment Outcome
17.
Trials ; 23(1): 789, 2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36127727

ABSTRACT

BACKGROUND: Mitochondrial disease is a heterogenous group of rare, complex neurometabolic disorders. Despite their individual rarity, collectively mitochondrial diseases represent the most common cause of inherited metabolic disorders in the UK; they affect 1 in every 4300 individuals, up to 15,000 adults (and a similar number of children) in the UK. Mitochondrial disease manifests multisystem and isolated organ involvement, commonly affecting those tissues with high energy demands, such as skeletal muscle. Myopathy manifesting as fatigue, muscle weakness and exercise intolerance is common and debilitating in patients with mitochondrial disease. Currently, there are no effective licensed treatments and consequently, there is an urgent clinical need to find an effective drug therapy. AIM: To investigate the efficacy of 12-week treatment with acipimox on the adenosine triphosphate (ATP) content of skeletal muscle in patients with mitochondrial disease and myopathy. METHODS: AIMM is a single-centre, double blind, placebo-controlled, adaptive designed trial, evaluating the efficacy of 12 weeks' administration of acipimox on skeletal muscle ATP content in patients with mitochondrial myopathy. Eligible patients will receive the trial investigational medicinal product (IMP), either acipimox or matched placebo. Participants will also be prescribed low dose aspirin as a non-investigational medical product (nIMP) in order to protect the blinding of the treatment assignment. Eighty to 120 participants will be recruited as required, with an interim analysis for sample size re-estimation and futility assessment being undertaken once the primary outcome for 50 participants has been obtained. Randomisation will be on a 1:1 basis, stratified by Fatigue Impact Scale (FIS) (dichotomised as < 40, ≥ 40). Participants will take part in the trial for up to 20 weeks, from screening visits through to follow-up at 16 weeks post randomisation. The primary outcome of change in ATP content in skeletal muscle and secondary outcomes relating to quality of life, perceived fatigue, disease burden, limb function, balance and walking, skeletal muscle analysis and symptom-limited cardiopulmonary fitness (optional) will be assessed between baseline and 12 weeks. DISCUSSION: The AIMM trial will investigate the effect of acipimox on modulating muscle ATP content and whether it can be repurposed as a new treatment for mitochondrial disease with myopathy. TRIAL REGISTRATION: EudraCT2018-002721-29 . Registered on 24 December 2018, ISRCTN 12895613. Registered on 03 January 2019, https://www.isrctn.com/search?q=aimm.


Subject(s)
Mitochondrial Myopathies , Muscular Diseases , Adult , Child , Humans , Adenosine Triphosphate , Aspirin/therapeutic use , Fatigue , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/drug therapy , Pyrazines , Quality of Life , Randomized Controlled Trials as Topic
18.
Biochim Biophys Acta ; 1797(6-7): 890-6, 2010.
Article in English | MEDLINE | ID: mdl-20138159

ABSTRACT

Mitochondrial diseases originate from mutations in mitochondrial or nuclear genes encoding for mitochondrial proteome. Neurogenic muscle weakness, ataxia and retinitis pigmentosa (NARP) syndrome is associated with the T8993G transversion in ATP6 gene which results in substitution at the very conservative site in the subunit 6 of mitochondrial ATP synthase. Defects in the mitochondrial respiratory chain and the ATPase are considered to be accompanied by changes in the generation of reactive oxygen species (ROS). This study aimed to elucidate effects of selenium on ROS and antioxidant system of NARP cybrid cells with 98% of T8993G mutation load. We found that selenium decreased ROS generation and increased the level and activity of antioxidant enzymes such as glutathione peroxidase (GPx) and thioredoxin reductase (TrxR). Therefore, we propose selenium to be a promising therapeutic agent not only in the case of NARP syndrome but also other diseases associated with mitochondrial dysfunctions and oxidative stress.


Subject(s)
Antioxidants/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Reactive Oxygen Species/metabolism , Selenium/pharmacology , Antioxidants/pharmacology , Catalase/metabolism , Cell Line, Tumor , DNA, Mitochondrial/genetics , Humans , Hybrid Cells , Mitochondria/genetics , Mitochondrial Myopathies/drug therapy , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Mutation, Missense , NF-E2-Related Factor 2/metabolism , Retinitis Pigmentosa/drug therapy , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Superoxide Dismutase/metabolism , Syndrome , Thioredoxin-Disulfide Reductase/metabolism
20.
BMJ Case Rep ; 14(1)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33431453

ABSTRACT

Mitochondrial diseases are rare, often go undiagnosed and can lead to devastating cascades of multisystem organ dysfunction. This report of a young woman with hearing loss and gestational diabetes illustrates a novel presentation of a cardiomyopathy caused by a previously described mutation in a mitochondrial gene, MT-TL1. She initially had biventricular heart dysfunction and ventricular arrhythmia that ultimately recovered with beta blockade and time. She continues to participate in sport without decline. It is important to keep mitochondrial diseases in the differential diagnosis and understand the testing and management strategies in order to provide the best patient care.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Cardiomyopathies/diagnosis , Mitochondrial Myopathies/diagnosis , RNA, Transfer, Leu/genetics , Tachycardia, Ventricular/genetics , Adult , Cardiomyopathies/complications , Cardiomyopathies/drug therapy , Cardiomyopathies/genetics , Coronary Angiography , DNA Mutational Analysis , Diagnosis, Differential , Echocardiography , Female , Genetic Testing , Heart Ventricles/diagnostic imaging , Heart Ventricles/physiopathology , Humans , Magnetic Resonance Imaging , Martial Arts/physiology , Mitochondrial Myopathies/complications , Mitochondrial Myopathies/drug therapy , Mitochondrial Myopathies/genetics , Mutation , Tachycardia, Ventricular/diagnosis , Treatment Outcome , Troponin/blood
SELECTION OF CITATIONS
SEARCH DETAIL