Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int J Mol Sci ; 25(7)2024 Mar 24.
Article in English | MEDLINE | ID: mdl-38612442

ABSTRACT

MELAS syndrome, characterized by mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes, represents a devastating mitochondrial disease, with the stroke-like episodes being its primary manifestation. Arginine supplementation has been used and recommended as a treatment for these acute attacks; however, insufficient evidence exists to support this treatment for MELAS. The mechanisms underlying the effect of arginine on MELAS pathophysiology remain unclear, although it is hypothesized that arginine could increase nitric oxide availability and, consequently, enhance blood supply to the brain. A more comprehensive understanding of these mechanisms is necessary to improve treatment strategies, such as dose and regimen adjustments; identify which patients could benefit the most; and establish potential markers for follow-up. This review aims to analyze the existing evidence concerning the mechanisms through which arginine supplementation impacts MELAS pathophysiology and provide the current scenario and perspectives for future investigations.


Subject(s)
Acidosis, Lactic , MELAS Syndrome , Stroke , Humans , MELAS Syndrome/drug therapy , Arginine/therapeutic use , Dietary Supplements
2.
Am J Med Genet A ; 194(3): e63461, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37953071

ABSTRACT

The MT-TL2 m.12315G>A pathogenic variant has previously been reported in five individuals with mild clinical phenotypes. Herein we report the case of a 5-year-old child with heteroplasmy for this variant who developed neurological regression and stroke-like episodes similar to those observed in mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS). Biochemical evaluation revealed depletion of arginine on plasma amino acid analysis and low z-scores for citrulline on untargeted plasma metabolomics analysis. These findings suggested that decreased availability of nitric oxide may have contributed to the stroke-like episodes. The use of intravenous arginine during stroke-like episodes and daily enteral L-citrulline supplementation normalized her biochemical values of arginine and citrulline. Untargeted plasma metabolomics showed the absence of nicotinamide and 1-methylnicotinamide, and plasma total glutathione levels were low; thus, nicotinamide riboside and N-acetylcysteine therapies were initiated. This report expands the phenotype associated with the rare mitochondrial variant MT-TL2 m.12315G>A to include neurological regression and a MELAS-like phenotype. Individuals with this variant should undergo in-depth biochemical analysis to include untargeted plasma metabolomics, plasma amino acids, and glutathione levels to help guide a targeted approach to treatment.


Subject(s)
Acidosis, Lactic , MELAS Syndrome , Mitochondrial Encephalomyopathies , Stroke , Child, Preschool , Female , Humans , Arginine/genetics , Citrulline , Glutathione/metabolism , MELAS Syndrome/diagnosis , MELAS Syndrome/genetics , MELAS Syndrome/complications , Nitric Oxide Donors/metabolism , Stroke/complications , Stroke/drug therapy
3.
Neuroradiology ; 66(3): 389-398, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38114794

ABSTRACT

PURPOSE: MELAS syndrome is a genetic disorder caused by mitochondrial DNA mutations. We previously described that MELAS patients had increased CSF glutamate and decreased CSF glutamine levels and that oral glutamine supplementation restores these values. Proton magnetic resonance spectroscopy (1H-MRS) allows the in vivo evaluation of brain metabolism. We aimed to compare 1H-MRS of MELAS patients with controls, the 1H-MRS after glutamine supplementation in the MELAS group, and investigate the association between 1H-MRS and CSF lactate, glutamate, and glutamine levels. METHODS: We conducted an observational case-control study and an open-label, single-cohort study with single-voxel MRS (TE 144/35 ms). We assessed the brain metabolism changes in the prefrontal (PFC) and parieto-occipital) cortex (POC) after oral glutamine supplementation in MELAS patients. MR spectra were analyzed with jMRUI software. RESULTS: Nine patients with MELAS syndrome (35.8 ± 3.2 years) and nine sex- and age-matched controls were recruited. Lactate/creatine levels were increased in MELAS patients in both PFC and POC (0.40 ± 0.05 vs. 0, p < 0.001; 0.32 ± 0.03 vs. 0, p < 0.001, respectively). No differences were observed between groups in glutamate and glutamine (Glx/creatine), either in PFC (p = 0.930) or POC (p = 0.310). No differences were observed after glutamine supplementation. A positive correlation was found between CSF lactate and lactate/creatine only in POC (0.85, p = 0.003). CONCLUSION: No significant metabolite changes were observed in the brains of MELAS patients after glutamine supplementation. While we found a positive correlation between lactate levels in CSF and 1H-MRS in MELAS patients, we could not monitor treatment response over short periods with this tool. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT04948138; initial release 24/06/2021; first patient enrolled on 1/07/2021. https://clinicaltrials.gov/ct2/show/NCT04948138.


Subject(s)
Glutamine , MELAS Syndrome , Humans , Glutamine/metabolism , MELAS Syndrome/diagnostic imaging , MELAS Syndrome/drug therapy , MELAS Syndrome/metabolism , Creatine/metabolism , Case-Control Studies , Cohort Studies , Magnetic Resonance Spectroscopy/methods , Glutamic Acid/metabolism , Proton Magnetic Resonance Spectroscopy/methods , Lactates , Dietary Supplements
4.
Brain Nerve ; 75(5): 523-525, 2023 May.
Article in Japanese | MEDLINE | ID: mdl-37194524

ABSTRACT

In MELAS, taurine modification defect in the anticodon of mitochondrial leucine tRNA causes codon translation failure. An investigator-started clinical trials of high-dose taurine therapy, that showed its efficacy in preventing stroke-like episodes, and improving the taurine modification rate. The drug was found to be safe. Taurine has been approved as a drug covered by public insurance for prevention of stroke-like episodes since 2019. Recently, L-arginine hydrochloride has also been approved for off-label use as a treatment for both acute and intermittent stages of stroke-like episodes.


Subject(s)
MELAS Syndrome , Stroke , Humans , MELAS Syndrome/drug therapy , MELAS Syndrome/genetics , MELAS Syndrome/complications , Stroke/etiology , Arginine , Taurine/therapeutic use , Mitochondria
5.
Eur J Neurol ; 30(2): 538-547, 2023 02.
Article in English | MEDLINE | ID: mdl-36334048

ABSTRACT

BACKGROUND AND PURPOSE: Mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes (MELAS) syndrome is a genetically heterogeneous disorder caused by mitochondrial DNA mutations. There are no disease-modifying therapies, and treatment remains mainly supportive. It has been shown previously that patients with MELAS syndrome have significantly increased cerebrospinal fluid (CSF) glutamate and significantly decreased CSF glutamine levels compared to controls. Glutamine has many metabolic fates in neurons and astrocytes, and the glutamate-glutamine cycle couples with many metabolic pathways depending on cellular requirements. The aim was to compare CSF glutamate and glutamine levels before and after dietary glutamine supplementation. It is postulated that high-dose oral glutamine supplementation could reduce the increase in glutamate levels. METHOD: This open-label, single-cohort study determined the safety and changes in glutamate and glutamine levels in CSF after 12 weeks of oral glutamine supplementation. RESULTS: Nine adult patients with MELAS syndrome (66.7% females, mean age 35.8 ± 3.2 years) were included. After glutamine supplementation, CSF glutamate levels were significantly reduced (9.77 ± 1.21 vs. 18.48 ± 1.34 µmol/l, p < 0.001) and CSF glutamine levels were significantly increased (433.66 ± 15.31 vs. 336.31 ± 12.92 µmol/l, p = 0.002). A side effect observed in four of nine patients was a mild sensation of satiety. One patient developed mild and transient elevation of transaminases, and another patient was admitted for an epileptic status without stroke-like episode. DISCUSSION: This study demonstrates that high-dose oral glutamine supplementation significantly reduces CSF glutamate and increases CSF glutamine levels in patients with MELAS syndrome. These findings may have potential therapeutic implications in these patients. TRIAL REGISTRATION INFORMATION: ClinicalTrials.gov Identifier: NCT04948138. Initial release 24 June 2021, first patient enrolled 1 July 2021. https://clinicaltrials.gov/ct2/show/NCT04948138.


Subject(s)
Acidosis, Lactic , MELAS Syndrome , Stroke , Adult , Female , Humans , Male , Cohort Studies , Dietary Supplements , Glutamic Acid/therapeutic use , Glutamine/therapeutic use , MELAS Syndrome/drug therapy , MELAS Syndrome/genetics , MELAS Syndrome/metabolism
6.
Stem Cell Res Ther ; 13(1): 260, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715869

ABSTRACT

BACKGROUND: Mitochondrial dysfunction and mitochondrial DNA (mtDNA) damage in the retinal pigment epithelium (RPE) have been implicated in the pathogenesis of age-related macular degeneration (AMD). However, a deeper understanding is required to determine the contribution of mitochondrial dysfunction and impaired mitochondrial autophagy (mitophagy) to RPE damage and AMD pathobiology. In this study, we model the impact of a prototypical systemic mitochondrial defect, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS), in RPE health and homeostasis as an in vitro model for impaired mitochondrial bioenergetics. METHODS: We used induced pluripotent stem cells (iPSCs) derived from skin biopsies of MELAS patients (m.3243A > G tRNA leu mutation) with different levels of mtDNA heteroplasmy and differentiated them into RPE cells. Mitochondrial depletion of ARPE-19 cells (p0 cells) was also performed using 50 ng/mL ethidium bromide (EtBr) and 50 mg/ml uridine. Cell fusion of the human platelets with the p0 cells performed using polyethylene glycol (PEG)/suspension essential medium (SMEM) mixture to generate platelet/RPE "cybrids." Confocal microscopy, FLowSight Imaging cytometry, and Seahorse XF Mito Stress test were used to analyze mitochondrial function. Western Blotting was used to analyze expression of autophagy and mitophagy proteins. RESULTS: We found that MELAS iPSC-derived RPE cells exhibited key characteristics of native RPE. We observed heteroplasmy-dependent impairment of mitochondrial bioenergetics and reliance on glycolysis for generating energy in the MELAS iPSC-derived RPE. The degree of heteroplasmy was directly associated with increased activation of signal transducer and activator of transcription 3 (STAT3), reduced adenosine monophosphate-activated protein kinase α (AMPKα) activation, and decreased autophagic activity. In addition, impaired autophagy was associated with aberrant lysosomal function, and failure of mitochondrial recycling. The mitochondria-depleted p0 cells replicated the effects on autophagy impairment and aberrant STAT3/AMPKα signaling and showed reduced mitochondrial respiration, demonstrating phenotypic similarities between p0 and MELAS iPSC-derived RPE cells. CONCLUSIONS: Our studies demonstrate that the MELAS iPSC-derived disease models are powerful tools for dissecting the molecular mechanisms by which mitochondrial DNA alterations influence RPE function in aging and macular degeneration, and for testing novel therapeutics in patients harboring the MELAS genotype.


Subject(s)
Induced Pluripotent Stem Cells , MELAS Syndrome , Macular Degeneration , Autophagy/genetics , DNA, Mitochondrial/genetics , Energy Metabolism/genetics , Epithelial Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , MELAS Syndrome/genetics , MELAS Syndrome/metabolism , MELAS Syndrome/pathology , Macular Degeneration/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigments/metabolism
7.
Brain Dev ; 44(8): 583-587, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35562277

ABSTRACT

BACKGROUND: Mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) with aphasia is a rare disorder, with the associated aphasia reported as either Wernicke's or Broca's. Herein, we report a patient with MELAS complicated by thalamic aphasia. CASE: A 15-year-old right-handed girl presented with headache, nausea, right homonymous hemianopsia, and aphasia. She could repeat words said by others, but had word-finding difficulty, paraphasia, and dysgraphia. Brain MRI revealed abnormal signals from the left occipital lobe to the temporal lobe and left thalamus, but Wernicke's area and Broca's area were not involved. Additionally, she had short stature, lactic acidosis, bilateral sensorineural hearing loss, and a maternal family history of diabetes and mild deafness. Based on clinical findings and the presence of a mitochondrial A3243G mutation, she was diagnosed with MELAS. With treatment, the brain MRI lesions disappeared and her symptoms improved. Her aphasia was classified as amnesic aphasia because she could repeat words, despite having word-finding difficulty, paraphasia, and dysgraphia. Based on MRI findings of a left thalamic lesion, we diagnosed her with thalamic aphasia. CONCLUSION: Thalamic aphasia may be caused by MELAS. Assessment of whether repetition is preserved is important for classifying aphasia.


Subject(s)
Acidosis, Lactic , Agraphia , Aphasia , MELAS Syndrome , Stroke , Acidosis, Lactic/complications , Adolescent , Aphasia/etiology , Female , Humans , MELAS Syndrome/complications , MELAS Syndrome/diagnosis , Mitochondrial Encephalomyopathies , Stroke/complications , Stroke/diagnostic imaging , Thalamus/diagnostic imaging
8.
Metab Brain Dis ; 36(8): 2181-2193, 2021 12.
Article in English | MEDLINE | ID: mdl-34118021

ABSTRACT

Mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes (MELAS) is a disease that should be considered as a differential diagnosis to acute ischemic stroke taking into account its onset pattern and neurological symptoms, which are similar to those of an ischemic stroke. Technological advancements in neuroimaging modalities have greatly facilitated differential diagnosis between stroke and MELAS on diagnostic imaging. Stroke-like episodes in MELAS have the following features: (1) symptoms are neurolocalized according to lesion site; (2) epileptic seizures are often present; (3) lesion distribution is inconsistent with vascular territory; (4) lesions are common in the posterior brain regions; (5) lesions continuously develop in adjacent sites over several weeks or months; (6) neurological symptoms and stroke-like lesions tend to be reversible, as presented on magnetic resonance imaging; (7) the rate of recurrence is high; and; (8) brain dysfunction and atrophy are slowly progressive. The m.3243ANG mutation in the MT-TL1 gene encoding the mitochondrial tRNALeu(UUR) is most commonly associated with MELAS. Although the precise pathophysiology is still unclear, one possible hypothesis for these episodes is a neuronal hyperexcitability theory, including neuron-astrocyte uncoupling. Supplementation, such as with L-arginine or taurine, has been proposed as preventive treatments for stroke-like episodes. As this disease is still untreatable and devastating, numerous drugs are being tested, and new gene therapies hold great promise for the future. This article contributes to the understanding of MELAS and its implications for clinical practice, by deepening their insight into the latest pathophysiological hypotheses and therapeutic developments.


Subject(s)
Ischemic Stroke , MELAS Syndrome , Stroke , Brain/pathology , Humans , MELAS Syndrome/diagnostic imaging , MELAS Syndrome/genetics , MELAS Syndrome/therapy , RNA, Transfer, Leu , Stroke/diagnostic imaging , Stroke/therapy
9.
Oxid Med Cell Longev ; 2019: 9537504, 2019.
Article in English | MEDLINE | ID: mdl-31249652

ABSTRACT

Wharton's jelly mesenchymal stem cells (WJMSCs) transfer healthy mitochondria to cells harboring a mitochondrial DNA (mtDNA) defect. Mitochondrial myopathy, encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) is one of the major subgroups of mitochondrial diseases, caused by the mt.3243A>G point mutation in the mitochondrial tRNALeu(UUR) gene. The specific aim of the study is to investigate whether WJMSCs exert therapeutic effect for mitochondrial dysfunction in cells of MELAS patient through donating healthy mitochondria. We herein demonstrate that WJMSCs transfer healthy mitochondria into rotenone-stressed fibroblasts of a MELAS patient, thereby eliminating mutation burden and rescuing mitochondrial functions. In the coculture system in vitro study, WJMSCs transferred healthy mitochondria to rotenone-stressed MELAS fibroblasts. By inhibiting actin polymerization to block tunneling nanotubes (TNTs), the WJMSC-conducted mitochondrial transfer was abrogated. After mitochondrial transfer, the mt.3243A>G mutation burden of MELAS fibroblasts was reduced to an undetectable level, with long-term retention. Sequencing results confirmed that the transferred mitochondria were donated from WJMSCs. Furthermore, mitochondrial transfer of WJMSCs to MELAS fibroblasts improves mitochondrial functions and cellular performance, including protein translation of respiratory complexes, ROS overexpression, mitochondrial membrane potential, mitochondrial morphology and bioenergetics, cell proliferation, mitochondrion-dependent viability, and apoptotic resistance. This study demonstrates that WJMSCs exert bioenergetic therapeutic effects through mitochondrial transfer. This finding paves the way for the development of innovative treatments for MELAS and other mitochondrial diseases.


Subject(s)
Energy Metabolism , Fibroblasts/transplantation , MELAS Syndrome/therapy , Mesenchymal Stem Cells/cytology , Mitochondria/transplantation , Mutation , Rotenone/adverse effects , Wharton Jelly/cytology , Cells, Cultured , Coculture Techniques , Fibroblasts/metabolism , Humans , MELAS Syndrome/chemically induced , MELAS Syndrome/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Reactive Oxygen Species/metabolism , Uncoupling Agents/adverse effects
10.
J Neurol Neurosurg Psychiatry ; 90(5): 529-536, 2019 05.
Article in English | MEDLINE | ID: mdl-29666206

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the efficacy and safety of high-dose taurine supplementation for prevention of stroke-like episodes of MELAS (mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes), a rare genetic disorder caused by point mutations in the mitochondrial DNA that lead to a taurine modification defect at the first anticodon nucleotide of mitochondrial tRNALeu(UUR), resulting in failure to decode codons accurately. METHODS: After the nationwide survey of MELAS, we conducted a multicentre, open-label, phase III trial in which 10 patients with recurrent stroke-like episodes received high-dose taurine (9 g or 12 g per day) for 52 weeks. The primary endpoint was the complete prevention of stroke-like episodes during the evaluation period. The taurine modification rate of mitochondrial tRNALeu(UUR) was measured before and after the trial. RESULTS: The proportion of patients who reached the primary endpoint (100% responder rate) was 60% (95% CI 26.2% to 87.8%). The 50% responder rate, that is, the number of patients achieving a 50% or greater reduction in frequency of stroke-like episodes, was 80% (95% CI 44.4% to 97.5%). Taurine reduced the annual relapse rate of stroke-like episodes from 2.22 to 0.72 (P=0.001). Five patients showed a significant increase in the taurine modification of mitochondrial tRNALeu(UUR) from peripheral blood leukocytes (P<0.05). No severe adverse events were associated with taurine. CONCLUSIONS: The current study demonstrates that oral taurine supplementation can effectively reduce the recurrence of stroke-like episodes and increase taurine modification in mitochondrial tRNALeu(UUR) in MELAS. TRIAL REGISTRATION NUMBER: UMIN000011908.


Subject(s)
Dietary Supplements , MELAS Syndrome/complications , Stroke/etiology , Stroke/prevention & control , Taurine/therapeutic use , Administration, Oral , Adolescent , Adult , Female , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
11.
Article in English | WPRIM | ID: wpr-764506

ABSTRACT

Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is a maternally inherited mitochondrial disorder of which m.3243A>G is the most commonly associated mutation, resulting in an inability to meet the energy requirements of various organs. MELAS poses a diagnostic challenge owing to its multiple organ involvement and great clinical variability due to its heteroplasmic nature. We report three cases from a family who were initially misdiagnosed with myasthenia gravis or undiagnosed. Although there is no optimal consensus treatment approach for patients with MELAS because of the disease's heterogeneity, our 21-year-long therapy regimen of l-arginine, l-carnitine, and coenzyme Q10 supplementation combined with dietary management appeared to provide noticeable protection from the symptoms and complications. Prompt early diagnosis is important, as optimal multidisciplinary management and early intervention may improve outcomes.


Subject(s)
Humans , Acidosis, Lactic , Arginine , Carnitine , Consensus , DNA, Mitochondrial , Early Diagnosis , Early Intervention, Educational , Follow-Up Studies , MELAS Syndrome , Mitochondrial Diseases , Myasthenia Gravis , Population Characteristics
12.
J Nutr ; 147(7): 1251-1257, 2017 07.
Article in English | MEDLINE | ID: mdl-28515163

ABSTRACT

Mitochondrial disorders result from dysfunctional mitochondria that are unable to generate sufficient energy to meet the needs of various organs. Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is one of the most frequent maternally inherited mitochondrial disorders. There is growing evidence that nitric oxide (NO) deficiency occurs in MELAS syndrome and results in impaired blood perfusion that contributes significantly to several complications in this disease. NO is synthesized from arginine by NO synthase, which catalyzes the conversion of arginine to NO and citrulline. Citrulline can be recycled into arginine, and therefore, both arginine and citrulline support NO synthesis. The use of 15N2-arginine and 13C-,2H4-citrulline stable isotope infusion allows measuring arginine flux; citrulline flux; citrulline-to-arginine flux, which represents the de novo arginine synthesis rate; and arginine-to-citrulline flux, which represents the NO production rate. The objective of this review is to highlight the utility of this method in providing additional evidence for NO deficiency in MELAS syndrome, adding more insight into the potential mechanisms of NO deficiency in this syndrome, and allowing for the assessment of the effects of supplementation with the NO donors, arginine and citrulline, on improving NO production in MELAS syndrome.


Subject(s)
MELAS Syndrome/metabolism , Nitric Oxide/metabolism , Arginine/metabolism , Citrulline/metabolism , Diagnostic Techniques, Radioisotope , Humans , Mitochondria/metabolism
13.
Acta neurol. colomb ; 33(1): 22-27, ene.-mar. 2017. graf
Article in Spanish | LILACS | ID: biblio-886418

ABSTRACT

RESUMEN Presentamos el caso clínico de una paciente adulta joven con episodios recurrentes sugestivos de ataque cerebrovascular, con cambios radiológicos típicos de enfermedad de MELAS con confirmación genética de mutación en el gen A3243G.


SUMMARY A clinical case of a young adult patient with recurrent episodes suggestive of stroke, with typical radiological changes of MELAS disease with genetic confirmation of mutation in A3243G gene.


Subject(s)
Spectrum Analysis , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , MELAS Syndrome , Equine-Assisted Therapy
14.
J Neuromuscul Dis ; 3(3): 363-379, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27854233

ABSTRACT

BACKGROUND: Mitochondrial encephalomyopathies are severe, relentlessly progressive conditions and there are very few effective therapies available to date. We have previously suggested that in two rare forms of reversible mitochondrial disease (reversible infantile respiratory chain deficiency and reversible infantile hepatopathy) supplementation with L-cysteine can improve mitochondrial protein synthesis, since cysteine is required for the 2-thiomodification of mitochondrial tRNAs. OBJECTIVES: We studied whether supplementation with L-cysteine or N-acetyl-cysteine (NAC) results in any improvement of the mitochondrial function in vitro in fibroblasts of patients with different genetic forms of abnormal mitochondrial translation. METHODS: We studied in vitro in fibroblasts of patients carrying the common m.3243A>G and m.8344A>G mutations or autosomal recessive mutations in genes affecting mitochondrial translation, whether L-cysteine or N-acetyl-cysteine supplementation have an effect on mitochondrial respiratory chain function. RESULTS: Here we show that supplementation with L-cysteine, but not with N-acetyl-cysteine partially rescues the mitochondrial translation defect in vitro in fibroblasts of patients carrying the m.3243A>G and m.8344A>G mutations. In contrast, N-acetyl-cysteine had a beneficial effect on mitochondrial translation in TRMU and MTO1 deficient fibroblasts. CONCLUSIONS: Our results suggest that L-cysteine or N-acetyl-cysteine supplementation may be a potential treatment for selected subgroups of patients with mitochondrial translation deficiencies. Further studies are needed to explore the full potential of cysteine supplementation as a treatment for patients with mitochondrial disease.


Subject(s)
Acetylcysteine/pharmacology , Cysteine/pharmacology , Fibroblasts/drug effects , MELAS Syndrome/metabolism , MERRF Syndrome/metabolism , Mitochondria/drug effects , Mitochondrial Diseases/metabolism , Protein Biosynthesis/drug effects , Carrier Proteins/genetics , Cyclooxygenase 2/genetics , Dietary Supplements , Fibroblasts/metabolism , Humans , In Vitro Techniques , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mutation , Neoplasm Proteins/genetics , Oxygen Consumption/drug effects , RNA-Binding Proteins , tRNA Methyltransferases/genetics
15.
PLoS One ; 11(4): e0154371, 2016.
Article in English | MEDLINE | ID: mdl-27110715

ABSTRACT

Mutations in mitochondrial DNA (mtDNA) can cause mitochondrial disease, a group of metabolic disorders that affect both children and adults. Interestingly, individual mtDNA mutations can cause very different clinical symptoms, however the factors that determine these phenotypes remain obscure. Defects in mitochondrial oxidative phosphorylation can disrupt cell signaling pathways, which may shape these disease phenotypes. In particular, mitochondria participate closely in cellular calcium signaling, with profound impact on cell function. Here, we examined the effects of a homoplasmic m.13565C>T mutation in MT-ND5 on cellular calcium handling using transmitochondrial cybrids (ND5 mutant cybrids). We found that the oxidation of NADH and mitochondrial membrane potential (Δψm) were significantly reduced in ND5 mutant cybrids. These metabolic defects were associated with a significant decrease in calcium uptake by ND5 mutant mitochondria in response to a calcium transient. Inhibition of glycolysis with 2-deoxy-D-glucose did not affect cytosolic calcium levels in control cybrids, but caused an increase in cytosolic calcium in ND5 mutant cybrids. This suggests that glycolytically-generated ATP is required not only to maintain Δψm in ND5 mutant mitochondria but is also critical for regulating cellular calcium homeostasis. We conclude that the m.13565C>T mutation in MT-ND5 causes defects in both mitochondrial oxidative metabolism and mitochondrial calcium sequestration. This disruption of mitochondrial calcium handling, which leads to defects in cellular calcium homeostasis, may be an important contributor to mitochondrial disease pathogenesis.


Subject(s)
Calcium/metabolism , Electron Transport Complex I/genetics , Fibroblasts/metabolism , Hybrid Cells/metabolism , MELAS Syndrome/genetics , Mitochondrial Proteins/genetics , Adenosine Triphosphate/biosynthesis , Cell Line, Tumor , Deoxyglucose/pharmacology , Electron Transport Complex I/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Regulation , Glycolysis/drug effects , Glycolysis/genetics , Humans , Hybrid Cells/drug effects , Hybrid Cells/pathology , MELAS Syndrome/metabolism , MELAS Syndrome/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/metabolism , Mutation , NAD/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/pathology , Oxidative Phosphorylation/drug effects , Primary Cell Culture , Signal Transduction
16.
Mol Genet Metab ; 117(4): 407-12, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26851065

ABSTRACT

Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is one of the most frequent maternally inherited mitochondrial disorders. The pathogenesis of this syndrome is not fully understood and believed to result from several interacting mechanisms including impaired mitochondrial energy production, microvasculature angiopathy, and nitric oxide (NO) deficiency. NO deficiency in MELAS syndrome is likely to be multifactorial in origin with the decreased availability of the NO precursors, arginine and citrulline, playing a major role. In this study we used stable isotope infusion techniques to assess NO production in children with MELAS syndrome and healthy pediatric controls. We also assessed the effect of oral arginine and citrulline supplementations on NO production in children with MELAS syndrome. When compared to control subjects, children with MELAS syndrome were found to have lower NO production, arginine flux, plasma arginine, and citrulline flux. In children with MELAS syndrome, arginine supplementation resulted in increased NO production, arginine flux, and arginine concentration. Citrulline supplementation resulted in a greater increase of these parameters. Additionally, citrulline supplementation was associated with a robust increase in citrulline concentration and flux and de novo arginine synthesis rate. The greater effect of citrulline in increasing NO production is due to its greater ability to increase arginine availability particularly in the intracellular compartment in which NO synthesis takes place. This study, which is the first one to assess NO metabolism in children with mitochondrial diseases, adds more evidence to the notion that NO deficiency occurs in MELAS syndrome, suggests a better effect for citrulline because of its greater role as NO precursor, and indicates that impaired NO production occurs in children as well as adults with MELAS syndrome. Thus, the initiation of treatment with NO precursors may be beneficial earlier in life. Controlled clinical trials to assess the therapeutic effects of arginine and citrulline on clinical complications of MELAS syndrome are needed.


Subject(s)
Arginine/administration & dosage , Citrulline/administration & dosage , Dietary Supplements , MELAS Syndrome/diet therapy , MELAS Syndrome/metabolism , Nitric Oxide/biosynthesis , Adolescent , Arginine/pharmacokinetics , Case-Control Studies , Child , Child, Preschool , Citrulline/pharmacokinetics , Female , Humans , MELAS Syndrome/diagnosis , Male , Treatment Outcome
18.
PLoS One ; 10(5): e0127066, 2015.
Article in English | MEDLINE | ID: mdl-25993630

ABSTRACT

OBJECTIVE: To study the effects of L-arginine (L-Arg) on total body aerobic capacity and muscle metabolism as assessed by (31)Phosphorus Magnetic Resonance Spectroscopy ((31)P-MRS) in patients with MELAS (Mitochondrial Encephalomyopathy with Lactic Acidosis and Stroke-like episodes) syndrome. METHODS: We performed a case control study in 3 MELAS siblings (m.3243A>G tRNA(leu(UUR)) in MTTL1 gene) with different % blood mutant mtDNA to evaluate total body maximal aerobic capacity (VO(2peak)) using graded cycle ergometry and muscle metabolism using 31P-MRS. We then ran a clinical trial pilot study in MELAS sibs to assess response of these parameters to single dose and a 6-week steady-state trial of oral L-Arginine. RESULTS: At baseline (no L-Arg), MELAS had lower serum Arg (p = 0.001). On 3(1)P-MRS muscle at rest, MELAS subjects had increased phosphocreatine (PCr) (p = 0.05), decreased ATP (p = 0.018), and decreased intracellular Mg(2+) (p = 0.0002) when compared to matched controls. With L-arginine therapy, the following trends were noted in MELAS siblings on cycle ergometry: (1) increase in mean % maximum work at anaerobic threshold (AT) (2) increase in % maximum heart rate at AT (3) small increase in VO(2peak). On (31)P-MRS the following mean trends were noted: (1) A blunted decrease in pH after exercise (less acidosis) (2) increase in Pi/PCr ratio (ADP) suggesting increased work capacity (3) a faster half time of PCr recovery (marker of mitochondrial activity) following 5 minutes of moderate intensity exercise (4) increase in torque. SIGNIFICANCE: These results suggest an improvement in aerobic capacity and muscle metabolism in MELAS subjects in response to supplementation with L-Arg. Intramyocellular hypomagnesemia is a novel finding that warrants further study. CLASSIFICATION OF EVIDENCE: Class III evidence that L-arginine improves aerobic capacity and muscle metabolism in MELAS subjects. TRIAL REGISTRATION: ClinicalTrials.gov NCT01603446.


Subject(s)
Arginine/therapeutic use , Exercise/physiology , MELAS Syndrome/drug therapy , MELAS Syndrome/metabolism , Muscles/metabolism , Adolescent , Arginine/pharmacology , Case-Control Studies , Dose-Response Relationship, Drug , Ergometry , Female , Humans , MELAS Syndrome/physiopathology , Magnetic Resonance Spectroscopy , Male , Muscles/drug effects , Neuroimaging , Phosphocreatine/analogs & derivatives , Phosphocreatine/metabolism , Rest/physiology , Young Adult
19.
Free Radic Biol Med ; 84: 161-170, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25843656

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a growth factor with pleiotropic effects on regulating lipid and glucose metabolism. Its expression is increased in skeletal muscle of mice and humans with mitochondrial disorders. However, the effects of FGF21 on skeletal muscle in response to mitochondrial respiratory chain deficiency are largely unknown. Here we demonstrate that the increased expression of FGF21 is a compensatory response to respiratory chain deficiency. The mRNA and protein levels of FGF21 were robustly raised in skeletal muscle from patients with mitochondrial myopathy or MELAS. The mammalian target of rapamycin (mTOR) phosphorylation levels and its downstream targets, Yin Yang 1 (YY1) and peroxisome proliferator-activated receptor γ, coactivator 1α (PGC-1α), were increased by FGF21 treatment in C2C12 myoblasts. Activation of the mTOR-YY1-PGC1α pathway by FGF21 in myoblasts regulated energy homeostasis as demonstrated by significant increases in intracellular ATP synthesis, oxygen consumption rate, activity of citrate synthase, glycolysis, mitochondrial DNA copy number, and induction of the expression of key energy metabolic genes. The effects of FGF21 on mitochondrial function required phosphoinositide 3-kinase (PI3K), which activates mTOR. Inhibition of PI3K, mTOR, YY1, and PGC-1α activities attenuated the stimulating effects of FGF21 on intracellular ATP levels and mitochondrial gene expression. Our findings revealed that mitochondrial respiratory chain deficiency elicited a compensatory response in skeletal muscle by increasing the FGF21 expression levels in muscle, which resulted in enhanced mitochondrial function through an mTOR-YY1-PGC1α-dependent pathway in skeletal muscle.


Subject(s)
Energy Metabolism , Fibroblast Growth Factors/metabolism , MELAS Syndrome/metabolism , Muscle, Skeletal/metabolism , Signal Transduction , Animals , Cell Line , Humans , Mice , Mitochondria, Muscle/metabolism , Oxidation-Reduction , Oxygen Consumption , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , YY1 Transcription Factor/metabolism
20.
Int J Biochem Cell Biol ; 48: 85-91, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24412347

ABSTRACT

Mitochondria are found in all nucleated human cells and generate most of the cellular energy. Mitochondrial disorders result from dysfunctional mitochondria that are unable to generate sufficient ATP to meet the energy needs of various organs. Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is a frequent maternally inherited mitochondrial disorder. There is growing evidence that nitric oxide (NO) deficiency occurs in MELAS syndrome and results in impaired blood perfusion that contributes significantly to several complications including stroke-like episodes, myopathy, and lactic acidosis. Both arginine and citrulline act as NO precursors and their administration results in increased NO production and hence can potentially have therapeutic utility in MELAS syndrome. Citrulline raises NO production to a greater extent than arginine, therefore, citrulline may have a better therapeutic effect. Controlled studies assessing the effects of arginine or citrulline supplementation on different clinical aspects of MELAS syndrome are needed.


Subject(s)
Arginine/therapeutic use , Citrulline/therapeutic use , MELAS Syndrome/drug therapy , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide/deficiency , Arginine/pharmacology , Citrulline/pharmacology , Humans , MELAS Syndrome/metabolism , Mitochondria/genetics , Nitric Oxide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL