Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Naunyn Schmiedebergs Arch Pharmacol ; 394(11): 2197-2222, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34596729

RESUMEN

Coenzyme Q10 (ubiquinone or CoQ10) is a lipid molecule that acts as an electron mobile carrier of the electron transport chain and also contains antioxidant properties. Supplementation of CoQ10 has been very useful to treat mitochondrial diseases. CoQ10 along with its synthetic analogue, idebenone, is used largely to treat various neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and Friedreich's ataxia and additional brain disease condition like autism, multiple sclerosis, epilepsy, depression, and bipolar disorder, which are related to mitochondrial impairment. In this article, we have reviewed numerous physiological functions of CoQ10 and the rationale for its use in clinical practice in different brain disorders.


Asunto(s)
Encefalopatías/tratamiento farmacológico , Enfermedades Mitocondriales/tratamiento farmacológico , Ubiquinona/análogos & derivados , Animales , Antioxidantes/farmacología , Encefalopatías/fisiopatología , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Enfermedades Mitocondriales/fisiopatología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/fisiopatología , Ubiquinona/farmacología
2.
Curr Opin Crit Care ; 26(4): 346-354, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32487844

RESUMEN

PURPOSE OF REVIEW: Mitochondrial dysfunction is associated with increased morbidity and mortality during and after critical illness. The concept of adaptive mitochondrial metabolic-bio-energetic downregulation rather than bio-energetic failure during the acute phase of critical illness has gained traction. As mitochondria are not able to utilize substrate during adaptive hibernation and aggressive feeding induces further harm, this condition has consequences for nutrition therapy. RECENT FINDINGS: Meeting resting energy expenditure in early critical illness is associated with enhanced oxidative stress and attenuation of autophagy, as is hyperglycemia. The negative effect of early high protein administration remains unclear, whereas fat appears bio-energetically inert. Although antioxidant micronutrients are essential to mitochondrial function, high-dosage studies of single vitamins (C and D) failed to show benefit. Convalescence probably requires increased micronutrient and macronutrient administration to aid anabolism and restore mitochondrial function, although robust data on requirements and actual intake are lacking. SUMMARY: Optimal nutrition therapy in the early phase of critical illness should avoid overfeeding and preserve (adaptive) mitochondrial function. Micronutrient supplementation probably requires a strategic cocktail instead of a high dosage of a single nutrient. Focus on identification of distinct metabolic phases to adapt nutrition during and after critical illness is essential.


Asunto(s)
Enfermedad Crítica , Enfermedades Mitocondriales , Terapia Nutricional , Estrés Fisiológico , Convalecencia , Humanos , Mitocondrias , Enfermedades Mitocondriales/fisiopatología
3.
Continuum (Minneap Minn) ; 25(6): 1732-1766, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31794469

RESUMEN

PURPOSE OF REVIEW: This article provides an overview of mitochondrial and metabolic biology, the genetic mechanisms causing mitochondrial diseases, the clinical features of mitochondrial diseases, lipid myopathies, and glycogen storage diseases, all with a focus on those syndromes and diseases associated with myopathy. Over the past decade, advances in genetic testing have revolutionized patient evaluation. The main goal of this review is to give the clinician the basic understanding to recognize patients at risk of these diseases using the standard history and physical examination. RECENT FINDINGS: Primary mitochondrial disease is the current designation for the illnesses resulting from genetic mutations in genes whose protein products are necessary for mitochondrial structure or function. In most circumstances, more than one organ system is involved in mitochondrial disease, and the value of the classic clinical features as originally described early in the history of mitochondrial diseases has reemerged as being important to identifying patients who may have a primary mitochondrial disease. The use of the genetic laboratory has become the most powerful tool for confirming a diagnosis, and nuances of using genetic results will be discussed in this article. Treatment for mitochondrial disease is symptomatic, with less emphasis on vitamin and supplement therapy than in the past. Clinical trials using pharmacologic agents are in progress, with the field attempting to define proper goals of treatment. Several standard accepted therapies exist for many of the metabolic myopathies. SUMMARY: Mitochondrial, lipid, and glycogen diseases are not uncommon causes of multisystem organ dysfunction, with the neurologic features, especially myopathy, occurring as a predominant feature. Early recognition requires basic knowledge of the varied clinical phenotypes before moving forward with a screening evaluation and possibly a genetic evaluation. Aside from a few specific diseases for which there are recommended interventions, treatment for the majority of these disorders remains symptomatic, with clinical trials currently in progress that will hopefully result in standard treatments.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno , Trastornos del Metabolismo de los Lípidos , Enfermedades Mitocondriales , Enfermedades Musculares , Adolescente , Anciano , Femenino , Enfermedad del Almacenamiento de Glucógeno/diagnóstico , Enfermedad del Almacenamiento de Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno/fisiopatología , Humanos , Trastornos del Metabolismo de los Lípidos/diagnóstico , Trastornos del Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/fisiopatología , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/fisiopatología , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Enfermedades Musculares/fisiopatología
4.
Parkinsonism Relat Disord ; 68: 8-16, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31621627

RESUMEN

Cerebellar ataxia is a hallmark of coenzyme Q10 (CoQ10) deficiency associated with COQ8A mutations. We present four patients, one with novel COQ8A pathogenic variants all with early, prominent handwriting impairment, dystonia and only mild ataxia. To better define the phenotypic spectrum and course of COQ8A disease, we review the clinical presentation and evolution in 47 reported cases. Individuals with COQ8A mutation display great clinical variability and unpredictable responses to CoQ10 supplementation. Onset is typically during infancy or childhood with ataxic features associated with developmental delay or regression. When disease onset is later in life, first symptoms can include: incoordination, epilepsy, tremor, and deterioration of writing. The natural history is characterized by a progression to a multisystem brain disease dominated by ataxia, with disease severity inversely correlated with age at onset. Six previously reported cases share with ours, a clinical phenotype characterized by slowly progressive or static writing difficulties, focal dystonia, and speech disorder, with only minimal ataxia. The combination of writing difficulty, dystonia and ataxia is a distinctive constellation that is reminiscent of a previously described clinical entity called Dystonia Ataxia Syndrome (DYTCA) and is an important clinical indicator of COQ8A mutations, even when ataxia is mild or absent.


Asunto(s)
Ataxia , Progresión de la Enfermedad , Trastornos Distónicos , Escritura Manual , Heterocigoto , Enfermedades Mitocondriales , Proteínas Mitocondriales/genética , Debilidad Muscular , Ubiquinona/deficiencia , Adulto , Ataxia/complicaciones , Ataxia/epidemiología , Ataxia/etiología , Ataxia/genética , Ataxia/fisiopatología , Niño , Trastornos Distónicos/epidemiología , Trastornos Distónicos/etiología , Trastornos Distónicos/genética , Trastornos Distónicos/fisiopatología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/epidemiología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/fisiopatología , Debilidad Muscular/complicaciones , Debilidad Muscular/epidemiología , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Ubiquinona/genética , Adulto Joven
5.
Int J Cardiol ; 266: 206-212, 2018 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-29887449

RESUMEN

BACKGROUND: Iron deficiency (ID), with or without anemia, is frequent in heart failure patients, and iron supplementation improves patient condition. However, the link between ID (independently of anemia) and cardiac function is poorly understood, but could be explained by an impaired mitochondrial metabolism. Our aim was to explore this hypothesis in a mouse model. METHODS AND RESULTS: We developed a mouse model of ID without anemia, using a blood withdrawal followed by 3-weeks low iron diet. ID was confirmed by low spleen, liver and heart iron contents and the repression of HAMP gene coding for hepcidin. ID was corrected by a single ferric carboxymaltose (FCM) injection (ID + FCM mice). Hemoglobin levels were similar in ID, ID + FCM and control mice. ID mice had impaired physical performances and left ventricular function (echocardiography). Mitochondrial complex I activity of cardiomyocytes was significantly decreased in ID mice, but not complexes II, III and IV activities. ID + FCM mice had improved physical performance, cardiac function and complex I activity compared to ID mice. Using BN-PAGE, we did not observe complex I disassembly, but a reduced quantity of the whole enzyme complex I in ID mice, that was restored in ID + FCM mice. CONCLUSIONS: ID, independently of anemia, is responsible for a decreased left ventricular function, through a reduction in mitochondrial complex I activity, probably secondary to a decrease in complex I quantity. These abnormalities are reversed after iron treatment, and may explain, at least in part, the benefit of iron supplementation in heart failure patients with ID.


Asunto(s)
Modelos Animales de Enfermedad , Complejo I de Transporte de Electrón/metabolismo , Deficiencias de Hierro , Mitocondrias/metabolismo , Resistencia Física/fisiología , Disfunción Ventricular Izquierda/metabolismo , Anemia Ferropénica , Animales , Complejo I de Transporte de Electrón/deficiencia , Hierro/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Bazo/metabolismo , Disfunción Ventricular Izquierda/fisiopatología
6.
Mol Genet Metab ; 125(1-2): 38-43, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29530532

RESUMEN

BACKGROUND: Coenzyme Q10 (CoQ10) is an important component of the mitochondrial respiratory chain (RC) and is critical for energy production. Although the prevalence of CoQ10 deficiency is still unknown, the general consensus is that the condition is under-diagnosed. The aim of this study was to retrospectively investigate CoQ10 deficiency in frozen muscle specimens in a cohort of ethnically diverse patients who received muscle biopsies for the investigation of a possible RC deficiency (RCD). METHODS: Muscle samples were homogenized whereby 600 ×g supernatants were used to analyze RC enzyme activities, followed by quantification of CoQ10 by stable isotope dilution liquid chromatography tandem mass spectrometry. The experimental group consisted of 156 patients of which 76 had enzymatically confirmed RCDs. To further assist in the diagnosis of CoQ10 deficiency in this cohort, we included sequencing of 18 selected nuclear genes involved with CoQ10 biogenesis in 26 patients with low CoQ10 concentration in muscle samples. RESULTS: Central 95% reference intervals (RI) were established for CoQ10 normalized to citrate synthase (CS) or protein. Nine patients were considered CoQ10 deficient when expressed against CS, while 12 were considered deficient when expressed against protein. In two of these patients the molecular genetic cause could be confirmed, of which one would not have been identified as CoQ10 deficient if expressed only against protein content. CONCLUSION: In this retrospective study, we report a central 95% reference interval for 600 ×g muscle supernatants prepared from frozen samples. The study reiterates the importance of including CoQ10 quantification as part of a diagnostic approach to study mitochondrial disease as it may complement respiratory chain enzyme assays with the possible identification of patients that may benefit from CoQ10 supplementation. However, the anomaly that only a few patients were identified as CoQ10 deficient against both markers (CS and protein), while the majority of patients where only CoQ10 deficient against one of the markers (and not the other), remains problematic. We therefore conclude from our data that, to prevent possibly not diagnosing a potential CoQ10 deficiency, the expression of CoQ10 levels in muscle on both CS as well as protein content should be considered.


Asunto(s)
Ataxia/diagnóstico , Metabolismo Energético/genética , Mitocondrias/genética , Enfermedades Mitocondriales/diagnóstico , Debilidad Muscular/diagnóstico , Ubiquinona/análogos & derivados , Ubiquinona/deficiencia , Adulto , Ataxia/metabolismo , Ataxia/fisiopatología , Células Cultivadas , Transporte de Electrón/genética , Femenino , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Debilidad Muscular/metabolismo , Debilidad Muscular/fisiopatología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Estudios Retrospectivos , Ubiquinona/genética , Ubiquinona/metabolismo
7.
Elife ; 72018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29402381

RESUMEN

Insulin resistance in muscle, adipocytes and liver is a gateway to a number of metabolic diseases. Here, we show a selective deficiency in mitochondrial coenzyme Q (CoQ) in insulin-resistant adipose and muscle tissue. This defect was observed in a range of in vitro insulin resistance models and adipose tissue from insulin-resistant humans and was concomitant with lower expression of mevalonate/CoQ biosynthesis pathway proteins in most models. Pharmacologic or genetic manipulations that decreased mitochondrial CoQ triggered mitochondrial oxidants and insulin resistance while CoQ supplementation in either insulin-resistant cell models or mice restored normal insulin sensitivity. Specifically, lowering of mitochondrial CoQ caused insulin resistance in adipocytes as a result of increased superoxide/hydrogen peroxide production via complex II. These data suggest that mitochondrial CoQ is a proximal driver of mitochondrial oxidants and insulin resistance, and that mechanisms that restore mitochondrial CoQ may be effective therapeutic targets for treating insulin resistance.


Asunto(s)
Tejido Adiposo/patología , Ataxia , Resistencia a la Insulina , Mitocondrias/patología , Enfermedades Mitocondriales/fisiopatología , Debilidad Muscular , Músculos/patología , Oxidantes/metabolismo , Ubiquinona/deficiencia , Adipocitos/fisiología , Animales , Humanos , Ratones , Sensibilidad y Especificidad
8.
PLoS One ; 12(9): e0184104, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28863176

RESUMEN

Mutations in PANK2 lead to neurodegeneration with brain iron accumulation. PANK2 has a role in the biosynthesis of coenzyme A (CoA) from dietary vitamin B5, but the neuropathological mechanism and reasons for iron accumulation remain unknown. In this study, atypical patient-derived fibroblasts were reprogrammed into induced pluripotent stem cells (iPSCs) and subsequently differentiated into cortical neuronal cells for studying disease mechanisms in human neurons. We observed no changes in PANK2 expression between control and patient cells, but a reduction in protein levels was apparent in patient cells. CoA homeostasis and cellular iron handling were normal, mitochondrial function was affected; displaying activated NADH-related and inhibited FADH-related respiration, resulting in increased mitochondrial membrane potential. This led to increased reactive oxygen species generation and lipid peroxidation in patient-derived neurons. These data suggest that mitochondrial deficiency is an early feature of the disease process and can be explained by altered NADH/FADH substrate supply to oxidative phosphorylation. Intriguingly, iron chelation appeared to exacerbate the mitochondrial phenotype in both control and patient neuronal cells. This raises caution for the use iron chelation therapy in general when iron accumulation is absent.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Enfermedades Mitocondriales/fisiopatología , Neurodegeneración Asociada a Pantotenato Quinasa/fisiopatología , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Acetilcoenzima A/química , Adolescente , Biopsia , Encéfalo/metabolismo , Diferenciación Celular , Niño , Coenzima A/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Hierro/química , Cariotipificación , Peroxidación de Lípido , Masculino , Potencial de la Membrana Mitocondrial , Mitocondrias/patología , Mutación , NAD/química , Neuronas/metabolismo , Ácido Pantoténico/química , Fenotipo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Plásmidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo
9.
EMBO Mol Med ; 9(1): 96-111, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27856618

RESUMEN

Coenzyme Q (CoQ) is an electron acceptor for sulfide-quinone reductase (SQR), the first enzyme of the hydrogen sulfide oxidation pathway. Here, we show that lack of CoQ in human skin fibroblasts causes impairment of hydrogen sulfide oxidation, proportional to the residual levels of CoQ. Biochemical and molecular abnormalities are rescued by CoQ supplementation in vitro and recapitulated by pharmacological inhibition of CoQ biosynthesis in skin fibroblasts and ADCK3 depletion in HeLa cells. Kidneys of Pdss2kd/kd mice, which only have ~15% residual CoQ concentrations and are clinically affected, showed (i) reduced protein levels of SQR and downstream enzymes, (ii) accumulation of hydrogen sulfides, and (iii) glutathione depletion. These abnormalities were not present in brain, which maintains ~30% residual CoQ and is clinically unaffected. In Pdss2kd/kd mice, we also observed low levels of plasma and urine thiosulfate and increased blood C4-C6 acylcarnitines. We propose that impairment of the sulfide oxidation pathway induced by decreased levels of CoQ causes accumulation of sulfides and consequent inhibition of short-chain acyl-CoA dehydrogenase and glutathione depletion, which contributes to increased oxidative stress and kidney failure.


Asunto(s)
Ataxia/fisiopatología , Enfermedades Mitocondriales/fisiopatología , Debilidad Muscular/fisiopatología , Sulfuros/metabolismo , Ubiquinona/deficiencia , Transferasas Alquil y Aril/deficiencia , Animales , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Oxidación-Reducción , Quinona Reductasas/análisis
10.
Biochim Biophys Acta ; 1857(8): 1079-1085, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27060254

RESUMEN

Coenzyme Q (CoQ, or ubiquinone) is a remarkable lipid that plays an essential role in mitochondria as an electron shuttle between complexes I and II of the respiratory chain, and complex III. It is also a cofactor of other dehydrogenases, a modulator of the permeability transition pore and an essential antioxidant. CoQ is synthesized in mitochondria by a set of at least 12 proteins that form a multiprotein complex. The exact composition of this complex is still unclear. Most of the genes involved in CoQ biosynthesis (COQ genes) have been studied in yeast and have mammalian orthologues. Some of them encode enzymes involved in the modification of the quinone ring of CoQ, but for others the precise function is unknown. Two genes appear to have a regulatory role: COQ8 (and its human counterparts ADCK3 and ADCK4) encodes a putative kinase, while PTC7 encodes a phosphatase required for the activation of Coq7. Mutations in human COQ genes cause primary CoQ(10) deficiency, a clinically heterogeneous mitochondrial disorder with onset from birth to the seventh decade, and with clinical manifestation ranging from fatal multisystem disorders, to isolated encephalopathy or nephropathy. The pathogenesis of CoQ(10) deficiency involves deficient ATP production and excessive ROS formation, but possibly other aspects of CoQ(10) function are implicated. CoQ(10) deficiency is unique among mitochondrial disorders since an effective treatment is available. Many patients respond to oral CoQ(10) supplementation. Nevertheless, treatment is still problematic because of the low bioavailability of the compound, and novel pharmacological approaches are currently being investigated. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.


Asunto(s)
Ataxia/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Debilidad Muscular/metabolismo , Ubiquinona/biosíntesis , Ubiquinona/deficiencia , Adenosina Trifosfato/agonistas , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/deficiencia , Animales , Ataxia/tratamiento farmacológico , Ataxia/genética , Ataxia/fisiopatología , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Humanos , Mitocondrias/genética , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/fisiopatología , Debilidad Muscular/tratamiento farmacológico , Debilidad Muscular/genética , Debilidad Muscular/fisiopatología , Mutación , Multimerización de Proteína , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Ubiquinona/genética , Ubiquinona/metabolismo , Ubiquinona/uso terapéutico
11.
Clin Genet ; 90(2): 156-60, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26818466

RESUMEN

Inherited ataxias are a group of heterogeneous disorders in children or adults but their genetic definition remains still undetermined in almost half of the patients. However, CoQ10 deficiency is a rare cause of cerebellar ataxia and ADCK3 is the most frequent gene associated with this defect. We herein report a 48 year old man, who presented with dysarthria and walking difficulties. Brain magnetic resonance imaging showed a marked cerebellar atrophy. Serum lactate was elevated. Tissues obtained by muscle and skin biopsies were studied for biochemical and genetic characterization. Skeletal muscle biochemistry revealed decreased activities of complexes I+III and II+III and a severe reduction of CoQ10 , while skin fibroblasts showed normal CoQ10 levels. A mild loss of maximal respiration capacity was also found by high-resolution respirometry. Molecular studies identified a novel homozygous deletion (c.504del_CT) in ADCK3, causing a premature stop codon. Western blot analysis revealed marked reduction of ADCK3 protein levels. Treatment with CoQ10 was started and, after 1 year follow-up, patient neurological condition slightly improved. This report suggests the importance of investigating mitochondrial function and, in particular, muscle CoQ10 levels, in patients with adult-onset cerebellar ataxia. Moreover, clinical stabilization by CoQ10 supplementation emphasizes the importance of an early diagnosis.


Asunto(s)
Ataxia/genética , Ataxia Cerebelosa/genética , Codón sin Sentido , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Enfermedades Mitocondriales/genética , Proteínas Mitocondriales/genética , Debilidad Muscular/genética , Ubiquinona/análogos & derivados , Ubiquinona/deficiencia , Ataxia/complicaciones , Ataxia/diagnóstico , Ataxia/fisiopatología , Ataxia Cerebelosa/complicaciones , Ataxia Cerebelosa/diagnóstico , Ataxia Cerebelosa/fisiopatología , Diagnóstico Tardío , Proteínas del Complejo de Cadena de Transporte de Electrón/deficiencia , Fibroblastos/metabolismo , Expresión Génica , Homocigoto , Humanos , Ácido Láctico/sangre , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/complicaciones , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/fisiopatología , Proteínas Mitocondriales/deficiencia , Debilidad Muscular/complicaciones , Debilidad Muscular/diagnóstico , Debilidad Muscular/fisiopatología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Piel/metabolismo , Ubiquinona/genética
12.
Drug Discov Today ; 20(11): 1349-54, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26183769

RESUMEN

Cellular folate metabolism is highly compartmentalized, with mitochondria folate transport and metabolism being distinct from the well-known cytosolic folate metabolism. There is evidence supporting the association between low folate status and mitochondrial DNA (mtDNA) instability, and cerebral folate deficiency is relatively frequent in mitochondrial disorders. Furthermore, folinic acid supplementation has been reported to be beneficial not only in some patients with mitochondrial disease, but also in patients with relatively common diseases where folate deficiency might be an important pathophysiological factor. In this review, we focus on the evidence that supports the potential involvement of impaired folate metabolism in the pathophysiology of mitochondrial disorders.


Asunto(s)
Deficiencia de Ácido Fólico/complicaciones , Ácido Fólico/metabolismo , Enfermedades Mitocondriales/fisiopatología , Animales , Transporte Biológico/fisiología , ADN Mitocondrial/metabolismo , Deficiencia de Ácido Fólico/tratamiento farmacológico , Humanos , Leucovorina/administración & dosificación , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/tratamiento farmacológico
13.
J Inherit Metab Dis ; 38(1): 145-56, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25091424

RESUMEN

Coenzyme Q(10) is a remarkable lipid involved in many cellular processes such as energy production through the mitochondrial respiratory chain (RC), beta-oxidation of fatty acids, and pyrimidine biosynthesis, but it is also one of the main cellular antioxidants. Its biosynthesis is still incompletely characterized and requires at least 15 genes. Mutations in eight of them (PDSS1, PDSS2, COQ2, COQ4, COQ6, ADCK3, ADCK4, and COQ9) cause primary CoQ(10) deficiency, a heterogeneous group of disorders with variable age of onset (from birth to the seventh decade) and associated clinical phenotypes, ranging from a fatal multisystem disease to isolated steroid resistant nephrotic syndrome (SRNS) or isolated central nervous system disease. The pathogenesis is complex and related to the different functions of CoQ(10). It involves defective ATP production and oxidative stress, but also an impairment of pyrimidine biosynthesis and increased apoptosis. CoQ(10) deficiency can also be observed in patients with defects unrelated to CoQ(10) biosynthesis, such as RC defects, multiple acyl-CoA dehydrogenase deficiency, and ataxia and oculomotor apraxia.Patients with both primary and secondary deficiencies benefit from high-dose oral supplementation with CoQ(10). In primary forms treatment can stop the progression of both SRNS and encephalopathy, hence the critical importance of a prompt diagnosis. Treatment may be beneficial also for secondary forms, although with less striking results.In this review we will focus on CoQ(10) biosynthesis in humans, on the genetic defects and the specific clinical phenotypes associated with CoQ(10) deficiency, and on the diagnostic strategies for these conditions.


Asunto(s)
Ataxia/diagnóstico , Ataxia/genética , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Debilidad Muscular/diagnóstico , Debilidad Muscular/genética , Ubiquinona/deficiencia , Adenosina Trifosfato/química , Animales , Ataxia/fisiopatología , Enfermedades del Sistema Nervioso Central/metabolismo , Modelos Animales de Enfermedad , Transporte de Electrón , Humanos , Ratones , Mitocondrias/metabolismo , Enfermedades Mitocondriales/fisiopatología , Debilidad Muscular/fisiopatología , Síndrome Nefrótico/metabolismo , Estrés Oxidativo , Fenotipo , Ubiquinona/análogos & derivados , Ubiquinona/química , Ubiquinona/genética
14.
J Trace Elem Med Biol ; 31: 157-62, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25511910

RESUMEN

A short review is given of the potential role of selenium deficiency and selenium intervention trials in atherosclerotic heart disease. Selenium is an essential constituent of several proteins, including the glutathione peroxidases and selenoprotein P. The selenium intake in Europe is generally in the lower margin of recommendations from authorities. Segments of populations in Europe may thus have a deficient intake that may be presented by a deficient anti-oxidative capacity in various illnesses, in particular atherosclerotic disease, and this may influence the prognosis of the disease. Ischemic heart disease and heart failure are two conditions where increased oxidative stress has been convincingly demonstrated. Some of the intervention studies of anti-oxidative substances that have focused on selenium are discussed in this review. The interrelationship between selenium and coenzyme Q10, another anti-oxidant, is presented, pointing to a theoretical advantage in using both substances in an intervention if there are deficiencies within the population. Clinical results from an intervention study using both selenium and coenzyme Q10 in an elderly population are discussed, where reduction in cardiovascular mortality, a better cardiac function according to echocardiography, and finally a lower concentration of the biomarker NT-proBNP as a sign of lower myocardial wall tension could be seen in those on active treatment, compared to placebo.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Enfermedades Carenciales/prevención & control , Dieta , Estado Nutricional , Estrés Oxidativo , Selenio/uso terapéutico , Ubiquinona/análogos & derivados , Animales , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Ataxia/metabolismo , Ataxia/fisiopatología , Ataxia/prevención & control , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/fisiopatología , Enfermedad de la Arteria Coronaria/prevención & control , Enfermedades Carenciales/metabolismo , Enfermedades Carenciales/fisiopatología , Dieta/efectos adversos , Suplementos Dietéticos , Europa (Continente) , Humanos , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Enfermedades Mitocondriales/prevención & control , Debilidad Muscular/metabolismo , Debilidad Muscular/fisiopatología , Debilidad Muscular/prevención & control , Selenio/deficiencia , Selenio/metabolismo , Ubiquinona/deficiencia , Ubiquinona/metabolismo , Ubiquinona/uso terapéutico
15.
Eur J Paediatr Neurol ; 18(4): 495-501, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24742911

RESUMEN

INTRODUCTION: There are many similarities, both clinical and radiological, between mitochondrial leukoencephalopathies and Alexander disease, an astrogliopathy. Clinically, both can manifest with a myriad of symptoms and signs, arising from the neonatal period to adulthood. Radiologically, both can demonstrate white matter changes, signal abnormalities of basal ganglia or thalami, brainstem abnormalities and contrast enhancement of white matter structures. Magnetic resonance spectroscopy may reveal elevation of lactate in the abnormal white matter in Alexander disease making the distinction even more challenging. PATIENT AND METHODS: We present a child who was considered to have an infantile onset mitochondrial disorder due to a combination of neurological symptoms and signs (developmental regression, failure to thrive, episodic deterioration, abnormal eye movements, pyramidal and cerebellar signs), urinary excretion of 3-methyl-glutaconic acid and imaging findings (extensive white matter changes and cerebellar atrophy) with a normal head circumference. Whole exome sequence analysis was performed. RESULTS: The child was found to harbor the R416W mutation, one of the most prevalent mutations in the glial fibrillary acidic protein (GFAP) gene that causes Alexander disease. CONCLUSIONS: Alexander disease should be considered in the differential diagnosis of infantile leukoencephalopathy, even when no macrocephaly is present. Next generation sequencing is a useful aid in unraveling the molecular etiology of leukoencephalopathies.


Asunto(s)
Enfermedad de Alexander/diagnóstico , Enfermedad de Alexander/genética , Exoma/genética , Proteína Ácida Fibrilar de la Glía/genética , Enfermedades Mitocondriales/fisiopatología , Mutación/genética , Arginina/genética , Preescolar , Femenino , Lóbulo Frontal/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Imagen por Resonancia Magnética , Triptófano/genética , Sustancia Blanca/patología
16.
Biochim Biophys Acta ; 1842(5): 677-85, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24530811

RESUMEN

Hypertrophic cardiomyopathy is a typical manifestation of very long-chain acyl-CoA dehydrogenase deficiency (VLCADD), the most common long-chain ß-oxidation defects in humans; however in some patients cardiac function is fully compensated. Cardiomyopathy may also be reversed by supplementation of medium-chain triglycerides (MCT). We here characterize cardiac function of VLCAD-deficient (VLCAD(-/-)) mice over one year. Furthermore, we investigate the long-term effect of a continuous MCT diet on the cardiac phenotype. We assessed cardiac morphology and function in VLCAD(-/-) mice by in vivo MRI. Cardiac energetics were measured by (31)P-MRS and myocardial glucose uptake was quantified by positron-emission-tomography (PET). Metabolic adaptations were identified by the expression of genes regulating glucose and lipid metabolism using real-time-PCR. VLCAD(-/-) mice showed a progressive decrease in heart function over 12 months accompanied by a reduced phosphocreatine-to-ATP-ratio indicative of chronic energy deficiency. Long-term MCT supplementation aggravated the cardiac phenotype into dilated cardiomyopathy with features similar to diabetic heart disease. Cardiac energy production and function in mice with a ß-oxidation defect cannot be maintained with age. Compensatory mechanisms are insufficient to preserve the cardiac energy state over time. However, energy deficiency by impaired ß-oxidation and long-term MCT induce cardiomyopathy by different mechanisms. Cardiac MRI and MRS may be excellent tools to assess minor changes in cardiac function and energetics in patients with ß-oxidation defects for preventive therapy.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga/deficiencia , Cardiomiopatías/fisiopatología , Errores Innatos del Metabolismo Lipídico/fisiopatología , Enfermedades Mitocondriales/fisiopatología , Enfermedades Musculares/fisiopatología , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Animales , Cardiomiopatías/genética , Síndromes Congénitos de Insuficiencia de la Médula Ósea , Metabolismo Energético , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Glucosa/metabolismo , Errores Innatos del Metabolismo Lipídico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Mitocondriales/genética , Enfermedades Musculares/genética , Miocardio/metabolismo , Reacción en Cadena de la Polimerasa , Tomografía de Emisión de Positrones , Sístole
18.
CNS Drugs ; 28(1): 63-8, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24242074

RESUMEN

BACKGROUND: Neurodegenerative movement disorders mainly include Parkinson's disease (PD), atypical parkinsonisms, Huntington's disease (HD), and Friedreich's ataxia (FA). With mitochondrial dysfunction observed in these diseases, mitochondrial enhancement such as creatine, coenzyme Q10 (CoQ10) and its analogues (idebenone and mitoquinone) has been regarded as a potential treatment. AIM: In this paper, we systematically analysed and summarized the efficacy of mitochondrial enhancement in improving motor and other symptoms in neurodegenerative movement disorders. METHODS: We searched the electronic databases PubMed, EMBASE, CINAHL, Cochrane Library and China National Knowledge Infrastructure until September 2013 for eligible randomized controlled trials (RCTs), as well as unpublished and ongoing trials. We calculated the mean differences for continuous data with 95% confidence intervals and pooled the results using a fixed-effect model, if no significant statistical heterogeneity was found (I(2) < 50%). RESULTS: We included 16 studies with 1,557 randomized patients, which compared creatine, CoQ10 or its analogues with placebo in motor and other symptoms. No significant improvements were found in the motor symptoms of PD, atypical parkinsonisms or HD patients, while only the high dose of idebenone seems to be promising for motor improvement in FA. Certain benefits are found in other symptoms. CONCLUSIONS: There is insufficient evidence to support the use of mitochondrial enhancement in patients with neurodegenerative movement disorders. More well-designed RCTs with large samples are required for further confirmation.


Asunto(s)
Creatina/uso terapéutico , Enfermedades Neurodegenerativas/tratamiento farmacológico , Ubiquinona/análogos & derivados , Animales , Creatina/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Compuestos Organofosforados/farmacología , Compuestos Organofosforados/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Ubiquinona/administración & dosificación , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
19.
J Surg Res ; 185(2): 805-14, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23899510

RESUMEN

BACKGROUND: Osthole, the main bioactive compounds isolated from the traditional Chinese medical herb broad Cnidium monnieri (L.) cusson, has been shown to exert spectrum of pharmacologic activities. The aim of this study was to investigate the potential neuroprotective effects of osthole against spinal cord ischemia-reperfusion injury in rats. MATERIALS AND METHODS: Osthole was administrated at the concentration of 0.1, 1, 10, 50, or 200 mg/kg (intraperitoneally) 1 h before spinal cord ischemia. The effects on spinal cord injury were measured by spinal cord water content, infarct volume, hematoxylin and eosin staining, and neurologic assessment. Mitochondria were purified from injured spinal cord tissue to determine mitochondrial function. RESULTS: We found that treatment with osthole (10 and 50 mg/kg) significantly decreased spinal cord water content and infarct volume, preserved normal motor neurons, and improved neurologic functions. These protective effects can be also observed even if the treatment was delayed to 4 h after reperfusion. Osthole treatment preserved mitochondrial membrane potential level, reduced reactive oxygen species production, increased adenosine triphosphate generation, and inhibited cytochrome c release in mitochondrial samples. Moreover, osthole increased mitochondria respiratory chain complex activities in spinal cord tissue, with no effect on mitochondrial DNA content and the expression of mitochondrial-specific transcription factors. CONCLUSIONS: All these findings demonstrate the neuroprotective effect of osthole in spinal cord ischemia-reperfusion injury model and suggest that oshtole-induced neuroprotection was mediated by mitochondrial biogenesis-independent inhibition of mitochondrial dysfunction.


Asunto(s)
Cumarinas/farmacología , Medicamentos Herbarios Chinos/farmacología , Enfermedades Mitocondriales/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/tratamiento farmacológico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Bloqueadores de los Canales de Calcio/farmacología , Cnidium/química , Hemodinámica/efectos de los fármacos , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/fisiopatología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología
20.
Clin Sci (Lond) ; 122(6): 289-97, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21970465

RESUMEN

MDs (mitochondrial diseases) are a clinically heterogeneous group of disorders characterized by impairment of the respiratory chain function with altered oxidative phosphorylation. We tested the hypothesis that the function of vascular endothelium is affected by increased oxidative stress in MDs. A total of 12 patients with MDs and pair-matched controls were studied. Endothelial function was assessed by measuring FMD (flow-mediated vasodilation) of brachial and common femoral arteries. The test was repeated after vitamin C (500 mg, twice a day) and E (400 mg, once a day) supplementation for 30 days and 90 days after vitamin withdrawal. FMD was reduced in patients compared with controls [AUC/τ (time-averaged area under the curve) for the brachial artery, 1.05±0.24 compared with 4.19±0.59% respectively, P<0.001; AUC/τ for the femoral artery, 0.98±0.19 compared with 2.36±0.29% respectively, P=0.001; values are means±S.E.M.] and correlated (brachial artery) with plasma lactate (r=-0.63, P<0.01). Urinary 8-iso-PGF2α (8-iso-prostaglandin F2α) was higher in patients than controls (505.6±85.9 compared with 302.5±38.7 pg/mg of creatinine; P<0.05) and correlated with plasma lactate (r=0.70, P<0.05). Immunohistochemical analysis showed 8-iso-PGF2α staining in MD-affected striated muscle cells and in blood vessels in muscle biopsies of patients. Antioxidant vitamins transiently restored FMD in patients [ΔAUC/τ (change in AUC/τ) for the brachial artery, +1.38±0.49%, P<0.05; ΔAUC/τ for the femoral artery, +0.98±0.24%, P<0.01] but had no effect on FMD in controls (brachial artery, -1.3±0.63%; and common femoral artery, -0.58±0.30%), thus abolishing the differences between patients and controls. The results of the present study indicate that oxidative stress is increased and is, at least partly, responsible for endothelial dysfunction in MDs.


Asunto(s)
Endotelio Vascular/fisiopatología , Enfermedades Mitocondriales/fisiopatología , Estrés Oxidativo , Adulto , Dinoprost/análogos & derivados , Dinoprost/orina , Femenino , Humanos , Masculino , Persona de Mediana Edad , Óxido Nítrico/fisiología , Especies Reactivas de Oxígeno , Vasodilatación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA