Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Eur J Clin Invest ; 54(7): e14217, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38644687

RESUMEN

OBJECTIVES AND SCOPE: Primary mitochondrial diseases (PMDs) are rare genetic disorders resulting from mutations in genes crucial for effective oxidative phosphorylation (OXPHOS) that can affect mitochondrial function. In this review, we examine the bioenergetic alterations and oxidative stress observed in cellular models of primary mitochondrial diseases (PMDs), shedding light on the intricate complexity between mitochondrial dysfunction and cellular pathology. We explore the diverse cellular models utilized to study PMDs, including patient-derived fibroblasts, induced pluripotent stem cells (iPSCs) and cybrids. Moreover, we also emphasize the connection between oxidative stress and neuroinflammation. INSIGHTS: The central nervous system (CNS) is particularly vulnerable to mitochondrial dysfunction due to its dependence on aerobic metabolism and the correct functioning of OXPHOS. Similar to other neurodegenerative diseases affecting the CNS, individuals with PMDs exhibit several neuroinflammatory hallmarks alongside neurodegeneration, a pattern also extensively observed in mouse models of mitochondrial diseases. Based on histopathological analysis of postmortem human brain tissue and findings in mouse models of PMDs, we posit that neuroinflammation is not merely a consequence of neurodegeneration but a potential pathogenic mechanism for disease progression that deserves further investigation. This recognition may pave the way for novel therapeutic strategies for this group of devastating diseases that currently lack effective treatments. SUMMARY: In summary, this review provides a comprehensive overview of bioenergetic alterations and redox imbalance in cellular models of PMDs while underscoring the significance of neuroinflammation as a potential driver in disease progression.


Asunto(s)
Metabolismo Energético , Enfermedades Mitocondriales , Enfermedades Neuroinflamatorias , Estrés Oxidativo , Humanos , Estrés Oxidativo/fisiología , Enfermedades Mitocondriales/fisiopatología , Enfermedades Mitocondriales/metabolismo , Enfermedades Neuroinflamatorias/fisiopatología , Enfermedades Neuroinflamatorias/metabolismo , Animales , Metabolismo Energético/fisiología , Fosforilación Oxidativa , Ratones , Mitocondrias/metabolismo , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , Síndrome MELAS/metabolismo , Síndrome MELAS/fisiopatología , Síndrome MELAS/genética , Modelos Animales de Enfermedad
2.
Ann Clin Transl Neurol ; 8(3): 704-710, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33476484

RESUMEN

The study aims to characterize the epilepsy phenotype of maternally inherited Leigh's syndrome (MILS) and neuropathy, ataxia, retinitis pigmentosa (NARP) due to mutations in the mitochondrial ATP6 gene and to correlate electroclinical features with mutant heteroplasmy load (HL). We investigated 17 individuals with different phenotype, from asymptomatic carriers to MILS: 11 carried the m.8993T> G mutation, 5 the m.8993T> C and one the novel, de novo m.8858G> A mutation. Seizures occurred in 37.5% of patients, EEG abnormalities in 73%. We ranked clinical and EEG abnormalities severity and performed quantitative EEG to estimate Abnormality Ratio (AR) and Spectral Relative Power (SRP). Spearman's rho and Kruskal-Wallis test were used for correlation with heteroplasmy load (HL). HL correlated with disease severity (Rho = 0.63, P = 0.012) and was significantly higher in patients with seizures or EEG abnormalities (P = 0.014). HL correlated with EEG severity score only for the m.8993T> G (Rho = 0.73, P = 0.040), showing a trend toward a positive correlation with AR and delta SPR, irrespective of the mutation.


Asunto(s)
Ondas Encefálicas/fisiología , Heteroplasmia/genética , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , Miopatías Mitocondriales/genética , Miopatías Mitocondriales/fisiopatología , ATPasas de Translocación de Protón Mitocondriales/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/fisiopatología , Adulto , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Linaje , Índice de Severidad de la Enfermedad
3.
Dev Med Child Neurol ; 63(6): 705-711, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33511646

RESUMEN

AIM: To investigate cerebral blood flow (CBF) in acute episodes of Leigh syndrome compared with basal state in patients carrying pathogenic mitochondrial disease gene variants responsible for neurometabolic disorders. METHOD: Arterial spin labelling (ASL) magnetic resonance imaging (MRI) sequences were used to measure CBF in 27 patients with mitochondrial respiratory chain enzyme deficiencies, ascribed to pathogenic variants of reported disease genes who were undergoing either urgent neuroimaging for acute episodes of Leigh syndrome (Group I: 15 MRI, seven females, eight males; mean age 7y; range 7mo-14y) or routine brain MRI (Group II: 15 MRI, eight females, seven males; mean age 5y 2mo; range 2mo-12y). RESULTS: Patients displayed markedly increased CBF in the striatum (2.8-fold greater, p<0.001 [1.05-2.53]) during acute episodes of Leigh syndrome compared to basal conditions. Detection of elevated CBF preceded identification of structural MRI lesions in four out of 15 cases. INTERPRETATION: Our results suggest that increased CBF is an overt hallmark of Leigh syndrome episodes and ASL MRI sequences should facilitate early diagnosis of acute episodes of Leigh syndrome, especially during the first attack in young children, when structural MRI is insufficiently informative.


Asunto(s)
Encéfalo/diagnóstico por imagen , Circulación Cerebrovascular/fisiología , Enfermedad de Leigh/diagnóstico por imagen , Enfermedades Mitocondriales/diagnóstico por imagen , Adolescente , Encéfalo/fisiopatología , Niño , Preescolar , Femenino , Humanos , Lactante , Enfermedad de Leigh/fisiopatología , Imagen por Resonancia Magnética , Masculino , Enfermedades Mitocondriales/fisiopatología , Marcadores de Spin
4.
Eur J Med Genet ; 63(10): 104003, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32679198

RESUMEN

Thiamine metabolism dysfunction syndrome (THMD) comprises a group of clinically and genetically heterogeneous encephalopathies with autosomal recessive inheritance. Four genes, SLC19A3, SLC25A19, SLC19A2, and TPK1, are associated with this disorder. This study aimed to explore the clinical, biochemical and molecular characteristics of seven Chinese patients with THMD. Targeted next-generation sequencing of mitochondrial DNA and nuclear DNA was used to identify the causative mutations. The patients presented with subacute encephalopathy between the ages of 1-27 months. Brain magnetic resonance imaging (MRI) revealed abnormalities in the basal ganglia, indicating Leigh syndrome. Urine α-ketoglutarate in five patients was elevated. In four patients, five novel mutations (c.1276_1278delTAC, c.265A > C, c.197T > C, c.850T > C, whole gene deletion) were found in SLC19A3, which is associated with THMD2. In two patients, four novel mutations (c.194C > T, c.454C > A, c.481G > A, and c.550G > C) were identified in SLC25A19, supporting a diagnosis of THMD4. In one patient, two novel mutations (c.395T > C and c.614-1G > A) were detected in TPK1, which is indicative of THMD5. The patients received thiamine, biotin, and symptomatic therapy, upon which six patients demonstrated clinical improvement. Our findings expanded the phenotypic and genotypic spectrum of THMD, with eleven novel mutations identified in seven Chinese patients. Early diagnosis and treatment have a significant impact on prognosis.


Asunto(s)
Encefalopatías/genética , ADN Mitocondrial/genética , Enfermedad de Leigh/genética , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Tiamina Pirofosfoquinasa/genética , Tiamina/metabolismo , Pueblo Asiatico , Biotina/uso terapéutico , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Encefalopatías/diagnóstico , Encefalopatías/diagnóstico por imagen , Encefalopatías/fisiopatología , Preescolar , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Ácidos Cetoglutáricos/orina , Enfermedad de Leigh/diagnóstico , Enfermedad de Leigh/diagnóstico por imagen , Enfermedad de Leigh/fisiopatología , Imagen por Resonancia Magnética , Masculino , Enfermedades Metabólicas/diagnóstico , Enfermedades Metabólicas/diagnóstico por imagen , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/fisiopatología , Tiamina/genética , Tiamina/uso terapéutico
5.
J Neuromuscul Dis ; 7(3): 301-308, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32444556

RESUMEN

BACKGROUND: In 2009, we identified TACO1 as a novel mitochondrial disease gene in a single family, however no second family has been described to confirm the role of TACO1 in mitochondrial disease. OBJECTIVE: In this report, we describe two independent consanguineous families carrying pathogenic variants in TACO1, confirming the phenotype. METHODS: Detailed clinical investigations and whole exome sequencing with haplotype analysis have been performed in several members of the two reported families. RESULTS: Clinical phenotype of the patients confirms the originally reported phenotype of a childhood-onset progressive cerebellar and pyramidal syndrome with optic atrophy and learning difficulties. Brain MRI showed periventricular white matter lesions with multiple cystic defects, suggesting leukoencephalopathy in both patients. One patient carried the previously described homozygous TACO1 variant (p.His158ProfsTer8) and haplotype analysis suggested that this variant is a rare founder mutation. The second patient from another family carried a homozygous novel frame shift variant (p.Cys85PhefsTer15). CONCLUSIONS: The identification of two Turkish families with similar characteristic clinical presentation and an additional homozygous nonsense mutation confirms that TACO1 is a human mitochondrial disease gene. Although most patients with this clinical presentation undergo next generation sequencing analysis, screening for selected founder mutations in the Turkish population based on the precise clinical presentation may reduce time and cost of finding the genetic diagnosis even in the era of massively parallel sequencing.


Asunto(s)
Enfermedad de Leigh/genética , Proteínas Mitocondriales/genética , Factores de Transcripción/genética , Adolescente , Adulto , Consanguinidad , Femenino , Humanos , Enfermedad de Leigh/diagnóstico por imagen , Enfermedad de Leigh/patología , Enfermedad de Leigh/fisiopatología , Masculino , Linaje , Turquía
6.
Elife ; 82019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31403401

RESUMEN

Mitochondrial deficits in energy production cause untreatable and fatal pathologies known as mitochondrial disease (MD). Central nervous system affectation is critical in Leigh Syndrome (LS), a common MD presentation, leading to motor and respiratory deficits, seizures and premature death. However, only specific neuronal populations are affected. Furthermore, their molecular identity and their contribution to the disease remains unknown. Here, using a mouse model of LS lacking the mitochondrial complex I subunit Ndufs4, we dissect the critical role of genetically-defined neuronal populations in LS progression. Ndufs4 inactivation in Vglut2-expressing glutamatergic neurons leads to decreased neuronal firing, brainstem inflammation, motor and respiratory deficits, and early death. In contrast, Ndufs4 deletion in GABAergic neurons causes basal ganglia inflammation without motor or respiratory involvement, but accompanied by hypothermia and severe epileptic seizures preceding death. These results provide novel insight in the cell type-specific contribution to the pathology, dissecting the underlying cellular mechanisms of MD.


Asunto(s)
Enfermedad de Leigh/patología , Enfermedad de Leigh/fisiopatología , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/fisiopatología , Neuronas/patología , Animales , Ganglios Basales/patología , Tronco Encefálico/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Complejo I de Transporte de Electrón/deficiencia , Ratones , Fenotipo
7.
Brain Dev ; 41(9): 803-807, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31178082

RESUMEN

An m.10158T>C mutation in MT-ND3, encoding a subunit of respiratory complex I, causes early-onset Leigh syndrome (LS), mitochondrial encephalomyopathy with lactic acid and stroke-like episodes (MELAS) syndrome, and LS and MELAS overlapping syndrome, presumably dependent on the ratio of heteroplasmy. Herein, we report a 4-year-old girl with heteroplasmic m.10158T>C mutation, showing an evolving age-dependent phenotype from LS to MELAS syndromes. She showed mild developmental delay during infancy, which was associated with magnetic resonance imaging lesions in the brain stem and basal ganglia. At the age of 4 years, she developed rapid neurological deterioration and intractable seizures, which was associated with recurrent multiple cerebral lesions as well as basal ganglia lesions. Her cerebral lesions were located predominantly in white matter and appeared at multiple areas simultaneously, unique characteristics that are distinct from typical MELAS. Two patients with LS-MELAS overlapping syndrome with m.10158T>C have been previously reported, however, this is the first patient with m.10158T>C showing significant age-dependent changes in clinical features and neuro-images, implying an age-dependent role of complex I in the developing brain.


Asunto(s)
Complejo I de Transporte de Electrón/genética , Enfermedad de Leigh/genética , Síndrome MELAS/genética , Mutación , Encéfalo/diagnóstico por imagen , Encéfalo/crecimiento & desarrollo , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Enfermedad de Leigh/diagnóstico por imagen , Enfermedad de Leigh/fisiopatología , Síndrome MELAS/diagnóstico por imagen , Síndrome MELAS/fisiopatología , Fenotipo
8.
J Hum Genet ; 64(4): 297-304, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30659264

RESUMEN

COQ4 mutations have recently been shown to cause a broad spectrum of mitochondrial disorders in association with CoQ10 deficiency. Herein, we report the clinical phenotype, in silico and biochemical analyses, and intervention for a novel c.370 G > A (p.G124S) COQ4 mutation in a Chinese family. This mutation is exclusively present in the East Asian population (allele frequency of ~0.001). The homozygous mutation caused CoQ10 deficiency-associated Leigh syndrome with an onset at 1-2 months of age, presenting as respiratory distress, lactic acidosis, dystonia, seizures, failure to thrive, and detectable lesions in the midbrain and basal ganglia. No renal impairment was involved. The levels of CoQ10 and mitochondrial respiratory chain complex (C) II + III activity were clearly lower in cultured fibroblasts derived from the patient than in those from unaffected carriers; the decreased CII + III activity could be increased by CoQ10 treatment. Follow-up studies suggested that our patient benefitted from the oral supplementation of CoQ10, which allowed her to maintain a relatively stable health status. Based on the genetic testing, preimplantation and prenatal diagnoses were performed, confirming that the next offspring of this family was unaffected. Our cases expand the phenotypic spectrum of COQ4 mutations and the genotypic spectrum of Leigh syndrome.


Asunto(s)
Ataxia/genética , Pruebas Genéticas , Enfermedad de Leigh/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Debilidad Muscular/genética , Ubiquinona/deficiencia , Pueblo Asiatico/genética , Ataxia/complicaciones , Preescolar , Simulación por Computador , Femenino , Fibroblastos/metabolismo , Heterocigoto , Homocigoto , Humanos , Lactante , Enfermedad de Leigh/complicaciones , Enfermedad de Leigh/fisiopatología , Masculino , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Mitocondriales/complicaciones , Debilidad Muscular/complicaciones , Mutación , Fenotipo , Ubiquinona/genética , Ubiquinona/farmacocinética
10.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L391-L399, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30520688

RESUMEN

Hypoxic pulmonary vasoconstriction (HPV) is a physiological vasomotor response that maintains systemic oxygenation by matching perfusion to ventilation during alveolar hypoxia. Although mitochondria appear to play an essential role in HPV, the impact of mitochondrial dysfunction on HPV remains incompletely defined. Mice lacking the mitochondrial complex I (CI) subunit Ndufs4 ( Ndufs4-/-) develop a fatal progressive encephalopathy and serve as a model for Leigh syndrome, the most common mitochondrial disease in children. Breathing normobaric 11% O2 prevents neurological disease and improves survival in Ndufs4-/- mice. In this study, we found that either genetic Ndufs4 deficiency or pharmacological inhibition of CI using piericidin A impaired the ability of left mainstem bronchus occlusion (LMBO) to induce HPV. In mice breathing air, the partial pressure of arterial oxygen during LMBO was lower in Ndufs4-/- and in piericidin A-treated Ndufs4+/+ mice than in respective controls. Impairment of HPV in Ndufs4-/- mice was not a result of nonspecific dysfunction of the pulmonary vascular contractile apparatus or pulmonary inflammation. In Ndufs4-deficient mice, 3 wk of breathing 11% O2 restored HPV in response to LMBO. When compared with Ndufs4-/- mice breathing air, chronic hypoxia improved systemic oxygenation during LMBO. The results of this study show that, when breathing air, mice with a congenital Ndufs4 deficiency or chemically inhibited CI function have impaired HPV. Our study raises the possibility that patients with inborn errors of mitochondrial function may also have defects in HPV.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Hipoxia/fisiopatología , Enfermedad de Leigh/fisiopatología , Vasoconstricción/fisiología , Animales , Bronquios/metabolismo , Modelos Animales de Enfermedad , Hipoxia/metabolismo , Pulmón/metabolismo , Pulmón/fisiopatología , Ratones Transgénicos , Mitocondrias/metabolismo , Arteria Pulmonar/metabolismo , Circulación Pulmonar/fisiología
11.
Proc Natl Acad Sci U S A ; 116(1): 277-286, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30578322

RESUMEN

The mitochondrial intramembrane rhomboid protease PARL has been implicated in diverse functions in vitro, but its physiological role in vivo remains unclear. Here we show that Parl ablation in mouse causes a necrotizing encephalomyelopathy similar to Leigh syndrome, a mitochondrial disease characterized by disrupted energy production. Mice with conditional PARL deficiency in the nervous system, but not in muscle, develop a similar phenotype as germline Parl KOs, demonstrating the vital role of PARL in neurological homeostasis. Genetic modification of two major PARL substrates, PINK1 and PGAM5, do not modify this severe neurological phenotype. Parl-/- brain mitochondria are affected by progressive ultrastructural changes and by defects in Complex III (CIII) activity, coenzyme Q (CoQ) biosynthesis, and mitochondrial calcium metabolism. PARL is necessary for the stable expression of TTC19, which is required for CIII activity, and of COQ4, which is essential in CoQ biosynthesis. Thus, PARL plays a previously overlooked constitutive role in the maintenance of the respiratory chain in the nervous system, and its deficiency causes progressive mitochondrial dysfunction and structural abnormalities leading to neuronal necrosis and Leigh-like syndrome.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Enfermedad de Leigh/etiología , Metaloproteasas/deficiencia , Proteínas Mitocondriales/deficiencia , Ubiquinona/metabolismo , Animales , Encéfalo/metabolismo , Calcio/metabolismo , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/fisiopatología , Hígado/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Encefalomiopatías Mitocondriales/metabolismo , Encefalomiopatías Mitocondriales/fisiopatología , Músculo Esquelético/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Endocr Regul ; 52(2): 110-118, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29715184

RESUMEN

OBJECTIVES: Leigh syndrome is a progressive early onset neurodegenerative disease typically presenting with psychomotor regression, signs of brainstem and/or basal ganglia disease, lactic acidosis, and characteristic magnetic resonance imaging findings. At molecular level, deficiency of respiratory complexes and/or pyruvate dehydrogenase complex is usually observed. Nuclear gene SURF1 encodes an assembly factor for cytochrome c-oxidase complex of the respiratory chain and autosomal recessive mutations in SURF1 are one of the most frequent causes of cytochrome c-oxidase-related Leigh syndrome cases. Here, we aimed to elucidate the genetic basis of Leigh syndrome in three Slovak families. METHODS AND RESULTS: Three probands presenting with Leigh syndrome were selected for DNA analysis. The first proband, presenting with atypical LS onset without abnormal basal ganglia magnetic resonance imaging findings, was analyzed with whole exome sequencing. In the two remaining probands, SURF1 was screened by Sanger sequencing. Four different heterozygous mutations were identified in SURF1: c.312_321delinsAT:p.(Pro104Profs*1), c.588+1G>A, c.823_833+7del:p. (?) and c.845_846del:p.(Ser282Cysfs*9). All the mutations are predicted to have a loss-of-function effect. CONCLUSIONS: We identified disease-causing mutations in all three probands, which points to the important role of SURF1 gene in etiology of Leigh syndrome in Slovakia. Our data showed that patients with atypical Leigh syndrome phenotype without lesions in basal ganglia may benefit from the whole exome sequencing method. In the case of probands presenting the typical phenotype, Sanger sequencing of the SURF1 gene seems to be an effective method of DNA analysis.


Asunto(s)
Enfermedad de Leigh/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Niño , Preescolar , Femenino , Humanos , Lactante , Enfermedad de Leigh/diagnóstico por imagen , Enfermedad de Leigh/patología , Enfermedad de Leigh/fisiopatología , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas Mitocondriales/antagonistas & inhibidores , Mutación , Linaje , Eslovaquia , Secuenciación del Exoma
13.
Artículo en Inglés | MEDLINE | ID: mdl-29686941

RESUMEN

Background: MEGDEL syndrome (3-MethylGlutaconic aciduria, Deafness, Encephalopathy, Leigh-like syndrome) is a severe neurometabolic disease with infantile onset. Phenomenology Shown: Progressive and marked dystonia over a 6-year period in an adult male with MEGDEL syndrome. Educational Value: Generalized dystonia may be the main manifestation of a milder form of MEGDEL syndrome, which begins during adulthood.


Asunto(s)
Encefalopatías/fisiopatología , Sordera/fisiopatología , Trastornos Distónicos/complicaciones , Enfermedad de Leigh/fisiopatología , Errores Innatos del Metabolismo/fisiopatología , Encéfalo/diagnóstico por imagen , Encefalopatías/diagnóstico por imagen , Sordera/diagnóstico por imagen , Progresión de la Enfermedad , Trastornos Distónicos/diagnóstico por imagen , Humanos , Enfermedad de Leigh/diagnóstico por imagen , Estudios Longitudinales , Masculino , Errores Innatos del Metabolismo/diagnóstico por imagen
14.
Mol Genet Metab ; 124(1): 71-81, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29602698

RESUMEN

In this study, we report a novel perpective of metabolic consequences for the m.8993T>G variant using fibroblasts from a proband with clinical symptoms compatible with Maternally Inherited Leigh Syndrome (MILS). Definitive diagnosis was corroborated by mitochondrial DNA testing for the pathogenic variant m.8993T>G in MT-ATP6 subunit by Sanger sequencing. The long-range PCR followed by massively parallel sequencing method detected the near homoplasmic m.8993T>G variant at 83% in the proband's fibroblasts and at 0.4% in the mother's fibroblasts. Our results are compatible with very low levels of germline heteroplasmy or an apparent de novo mutation. Our mitochondrial morphometric analysis reveals severe defects in mitochondrial cristae structure in the proband's fibroblasts. Our live-cell mitochondrial respiratory analyses show impaired oxidative phosphorylation with decreased spare respiratory capacity in response to energy stress in the proband's fibroblasts. We detected a diminished glycolysis with a lessened glycolytic capacity and reserve, revealing a stunted ability to switch to glycolysis upon full inhibition of OXPHOS activities. This dysregulated energy reprogramming results in a defective interplay between OXPHOS and glycolysis during an energy crisis. Our study sheds light on the potential pathophysiologic mechanism leading to chronic energy crisis in this MILS patient harboring the m.8993T>G variant.


Asunto(s)
Fibroblastos/metabolismo , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , ATPasas de Translocación de Protón Mitocondriales/genética , ADN Mitocondrial/genética , Metabolismo Energético , Femenino , Fibroblastos/citología , Glucólisis , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Enfermedad de Leigh/diagnóstico , Masculino , Mitocondrias/metabolismo , Madres , Mutación , Fosforilación Oxidativa , Linaje , Adulto Joven
15.
Hum Mol Genet ; 27(10): 1743-1753, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29518248

RESUMEN

LonP1 is a mitochondrial matrix protease whose selective substrate specificity is essential for maintaining mitochondrial homeostasis. Recessively inherited, pathogenic defects in LonP1 have been previously reported to underlie cerebral, ocular, dental, auricular and skeletal anomalies (CODAS) syndrome, a complex multisystemic and developmental disorder. Intriguingly, although classical mitochondrial disease presentations are well-known to exhibit marked clinical heterogeneity, the skeletal and dental features associated with CODAS syndrome are pathognomonic. We have applied whole exome sequencing to a patient with congenital lactic acidosis, muscle weakness, profound deficiencies in mitochondrial oxidative phosphorylation associated with loss of mtDNA copy number and MRI abnormalities consistent with Leigh syndrome, identifying biallelic variants in the LONP1 (NM_004793.3) gene; c.1693T > C predicting p.(Tyr565His) and c.2197G > A predicting p.(Glu733Lys); no evidence of the classical skeletal or dental defects observed in CODAS syndrome patients were noted in our patient. In vitro experiments confirmed the p.(Tyr565His) LonP1 mutant alone could not bind or degrade a substrate, consistent with the predicted function of Tyr565, whilst a second missense [p.(Glu733Lys)] variant had minimal effect. Mixtures of p.(Tyr565His) mutant and wild-type LonP1 retained partial protease activity but this was severely depleted when the p.(Tyr565His) mutant was mixed with the p.(Glu733Lys) mutant, data consistent with the compound heterozygosity detected in our patient. In summary, we conclude that pathogenic LONP1 variants can lead to a classical mitochondrial disease presentations associated with severe biochemical defects in oxidative phosphorylation in clinically relevant tissues.


Asunto(s)
Proteasas ATP-Dependientes/genética , Anomalías Craneofaciales/genética , Anomalías del Ojo/genética , Trastornos del Crecimiento/genética , Luxación Congénita de la Cadera/genética , Enfermedad de Leigh/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Osteocondrodisplasias/genética , Anomalías Dentarias/genética , Biopsia , Línea Celular , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/fisiopatología , Exoma/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/fisiopatología , Trastornos del Crecimiento/metabolismo , Trastornos del Crecimiento/fisiopatología , Luxación Congénita de la Cadera/metabolismo , Luxación Congénita de la Cadera/fisiopatología , Humanos , Lactante , Enfermedad de Leigh/metabolismo , Enfermedad de Leigh/fisiopatología , Masculino , Mitocondrias/genética , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Músculo Esquelético/fisiopatología , Mutación , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/fisiopatología , Fosforilación Oxidativa , Anomalías Dentarias/metabolismo , Anomalías Dentarias/fisiopatología , Secuenciación del Exoma
16.
Brain Dev ; 40(2): 145-149, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28916229

RESUMEN

BACKGROUND: Leigh syndrome is a mitochondrial disease caused by respiratory chain deficiency, and there are no proven effective therapies. EPI-743 is a potent cellular oxidative stress protectant and results of clinical trials for mitochondrial diseases are accumulating. CASE: At 5months, a girl presented with the scarce eye movement and diminished muscle tone. She was diagnosed with Leigh encephalopathy from blood and cerebrospinal fluid lactate elevation and MRI findings. Sequence analysis for mitochondrial DNA revealed a T10158C mutation in the mitochondrial encoded ND3 gene in complex I. RESULTS: At 8months, succinate was prescribed expected to restore the electron transport chain system. After that her condition got worse and succinate was discontinued. Subsequent administration of EPI-743 improved her eye movement, fine motor movements of the extremities, and bowel movement. She is now 5years old. Although brain atrophy has progressed, she has still respiratory free time. CONCLUSION: Our patient showed visible improvement with EPI-743 treatment and the only patient surviving after 4years. There is a possibility that EPI-743 is modifying the natural course of the syndrome.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad de Leigh/tratamiento farmacológico , Ubiquinona/análogos & derivados , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Preescolar , Progresión de la Enfermedad , Complejo I de Transporte de Electrón/genética , Femenino , Humanos , Ácido Láctico/sangre , Enfermedad de Leigh/diagnóstico por imagen , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , Respiración Artificial , Ubiquinona/uso terapéutico
17.
Clin Genet ; 93(2): 396-400, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28857146

RESUMEN

Deficiencies of mitochondrial respiratory chain complex I frequently result in leukoencephalopathy in young patients, and different mutations in the genes encoding its subunits are still being uncovered. We report 2 patients with cystic leukoencephalopathy and complex I deficiency with recessive mutations in NDUFA2, an accessory subunit of complex I. The first patient was initially diagnosed with a primary systemic carnitine deficiency associated with a homozygous variant in SLC22A5, but also exhibited developmental regression and cystic leukoencephalopathy, and an additional diagnosis of complex I deficiency was suspected. Biochemical analysis confirmed a complex I deficiency, and whole-exome sequencing revealed a homozygous mutation in NDUFA2 (c.134A>C, p.Lys45Thr). Review of a biorepository of patients with unsolved genetic leukoencephalopathies who underwent whole-exome or genome sequencing allowed us to identify a second patient with compound heterozygous mutations in NDUFA2 (c.134A>C, p.Lys45Thr; c.225del, p.Asn76Metfs*4). Only 1 other patient with mutations in NDUFA2 and a different phenotype (Leigh syndrome) has previously been reported. This is the first report of cystic leukoencephalopathy caused by mutations in NDUFA2.


Asunto(s)
Secuenciación del Exoma , Leucoencefalopatías/genética , Mitocondrias/genética , NADH Deshidrogenasa/genética , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Lactante , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , Leucoencefalopatías/fisiopatología , Masculino , Mitocondrias/patología , Mutación , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética
18.
Biochem Biophys Res Commun ; 493(1): 207-212, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28899781

RESUMEN

Mitochondrial diseases are the result of aberrant mitochondrial function caused by mutations in either nuclear or mitochondrial DNA. Poor bone health has recently been suggested as a symptom of mitochondrial diseases; however, a direct link between decreased mitochondrial function and poor bone health in mitochondrial disease has not been demonstrated. In this study, stem cells from human exfoliated deciduous teeth (SHED) were isolated from a child with Leigh syndrome (LS), a mitochondrial disease, and the effects of decreased mitochondrial function on poor bone health were analyzed. Compared with control SHED, LS SHED displayed decreased osteoblastic differentiation and calcium mineralization. The intracellular and mitochondrial calcium levels were lower in LS SHED than in control SHED. Furthermore, the mitochondrial activity of LS SHED was decreased compared with control SHED both with and without osteoblastic differentiation. Our results indicate that decreased osteoblast differentiation potential and osteoblast function contribute to poor bone health in mitochondrial diseases.


Asunto(s)
Calcio/metabolismo , Enfermedad de Leigh/fisiopatología , Mitocondrias/patología , Osteoblastos/patología , Células Madre/metabolismo , Células Madre/patología , Diente Primario/fisiopatología , Calcificación Fisiológica , Diferenciación Celular , Células Cultivadas , Niño , Preescolar , Femenino , Humanos , Enfermedad de Leigh/patología , Masculino , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Osteogénesis , Diente Primario/patología
19.
Mitochondrion ; 37: 62-79, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28739512

RESUMEN

The mitochondrial respiratory chain, and in particular, complex I, is a major source of reactive oxygen species (ROS) in cells. Elevated levels of ROS are associated with an imbalance between the rate of ROS formation and the capacity of the antioxidant defense system. Increased ROS production may lead to oxidation of DNA, lipids and proteins and thus can affect fundamental cellular processes. The aim of this study was to investigate the magnitude of intracellular oxidative stress in fibroblasts of patients with Leigh syndrome with defined mutations in complex I. Moreover, we hypothesized that activation of the p66Shc protein (phosphorylation of p66Shc at Ser36 by PKCß), being part of the oxidative stress response pathway, is partially responsible for the increased ROS production in cells with dysfunctional complex I. Characterization of bioenergetic parameters and ROS production showed that the cellular model of Leigh syndrome is described by increased intracellular oxidative stress and oxidative damage to DNA and proteins, which correlate with increased p66Shc phosphorylation at Ser36. Treatment of patients' fibroblasts with hispidin (an inhibitor of the protein kinase PKCß), in addition to decreasing ROS production and intracellular oxidative stress, resulted in restoration of complex I activity.


Asunto(s)
Fibroblastos/metabolismo , Enfermedad de Leigh/fisiopatología , Mitocondrias/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/antagonistas & inhibidores , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Células Cultivadas , Complejo I de Transporte de Electrón/genética , Inhibidores Enzimáticos/metabolismo , Humanos , Mutación , Pironas/metabolismo
20.
Med Hypotheses ; 101: 23-27, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28351484

RESUMEN

Leigh syndrome (LS) is a neurogenetic disorder of children caused by mutations in at least 75 genes which impair mitochondrial bioenergetics. The changes have typical localization in basal ganglia and brainstem, and typical histological picture of spongiform appearance, vascular proliferation and gliosis. ATP deprivation, free radicals and lactate accumulation are suspected to be the causes. Hypocapnic hypothesis proposed in the paper questions the energy deprivation as the mechanism of LS. We assume that the primary harmful factor is hypocapnia (decrease in pCO2) and respiratory alkalosis (increase in pH) due to hyperventilation, permanent or in response to stress. Inside mitochondria, the pH signal of high pH/low bicarbonate ion (HCO-3) is transmitted by soluble adenyl cyclase (sAC) through cAMP dependent manner. The process can initiate brain lesions (necrosis, apoptosis, hypervascularity) in OXPHOS deficient cells residing at the LS area of the brain. The major message of the article is that it is not the ATP depletion but intracellular alkalization (and/or hyperoxia?) which seem to be the cause of LS. The paper includes suggestions concerning the methodology for further research on the LS mechanism and for therapeutic strategy.


Asunto(s)
Hipocapnia/fisiopatología , Enfermedad de Leigh/terapia , Acidosis , Adenosina Trifosfato/química , Alcalosis Respiratoria/patología , Animales , Ganglios Basales/fisiopatología , Bicarbonatos/química , Análisis de los Gases de la Sangre , Lesiones Encefálicas/patología , Tronco Encefálico/fisiopatología , Dióxido de Carbono/química , Niño , AMP Cíclico/metabolismo , Radicales Libres/química , Gliosis/fisiopatología , Humanos , Concentración de Iones de Hidrógeno , Hiperventilación , Hipocapnia/genética , Hipoxia/patología , Lactatos/química , Ácido Láctico/química , Enfermedad de Leigh/genética , Enfermedad de Leigh/fisiopatología , Ratones , Mutación , Presión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA