Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell Proteomics ; 22(12): 100666, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37839702

RESUMEN

The application of integrated systems biology to the field of structural biology is a promising new direction, although it is still in the infant stages of development. Here we report the use of single particle cryo-EM to identify multiple proteins from three enriched heterogeneous fractions prepared from human liver mitochondrial lysate. We simultaneously identify and solve high-resolution structures of nine essential mitochondrial enzymes with key metabolic functions, including fatty acid catabolism, reactive oxidative species clearance, and amino acid metabolism. Our methodology also identified multiple distinct members of the acyl-CoA dehydrogenase family. This work highlights the potential of cryo-EM to explore tissue proteomics at the atomic level.


Asunto(s)
Mitocondrias , Proteómica , Humanos , Mitocondrias/metabolismo , Hígado/metabolismo , Oxidación-Reducción
2.
J Biol Chem ; 297(4): 101196, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34529976

RESUMEN

Mitochondria undergo continuous cycles of fission and fusion to promote inheritance, regulate quality control, and mitigate organelle stress. More recently, this process of mitochondrial dynamics has been demonstrated to be highly sensitive to nutrient supply, ultimately conferring bioenergetic plasticity to the organelle. However, whether regulators of mitochondrial dynamics play a causative role in nutrient regulation remains unclear. In this study, we generated a cellular loss-of-function model for dynamin-related protein 1 (DRP1), the primary regulator of outer membrane mitochondrial fission. Loss of DRP1 (shDRP1) resulted in extensive ultrastructural and functional remodeling of mitochondria, characterized by pleomorphic enlargement, increased electron density of the matrix, and defective NADH and succinate oxidation. Despite increased mitochondrial size and volume, shDRP1 cells exhibited reduced cellular glucose uptake and mitochondrial fatty acid oxidation. Untargeted transcriptomic profiling revealed severe downregulation of genes required for cellular and mitochondrial calcium homeostasis, which was coupled to loss of ATP-stimulated calcium flux and impaired substrate oxidation stimulated by exogenous calcium. The insights obtained herein suggest that DRP1 regulates substrate oxidation by altering whole-cell and mitochondrial calcium dynamics. These findings are relevant to the targetability of mitochondrial fission and have clinical relevance in the identification of treatments for fission-related pathologies such as hereditary neuropathies, inborn errors in metabolism, cancer, and chronic diseases.


Asunto(s)
Señalización del Calcio , Dinaminas/metabolismo , Mitocondrias Musculares/metabolismo , Dinámicas Mitocondriales , Línea Celular , Dinaminas/genética , Ácidos Grasos/genética , Ácidos Grasos/metabolismo , Humanos , Mitocondrias Musculares/genética , Oxidación-Reducción
3.
Clin Infect Dis ; 73(3): e765-e772, 2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-33564870

RESUMEN

BACKGROUND: Neurocognitive impairment (NCI) is associated with monocyte activation in people with HIV (PWH). Activated monocytes increase glycolysis, reduce oxidative phosphorylation, and accumulate citrate and succinate, tricarboxylic acid (TCA) cycle metabolites that promote inflammation-this metabolic shift may contribute to NCI and slowed gait speed in PWH. METHODS: Plasma citrate and succinate were assayed by liquid chromatography-mass spectrometry from 957 participants upon entry to a multicenter, prospective cohort of older PWH. Logistic, linear, and mixed-effects linear regression models were used to examine associations between entry/baseline TCA cycle metabolites and cross-sectional and longitudinal NCI, neuropsychological test scores (NPZ-4), and gait speed. RESULTS: Median age was 51 (range 40-78) years. Each 1 standard deviation (SD) citrate increment was associated with 1.18 higher odds of prevalent NCI at baseline (P = .03), 0.07 SD lower time-updated NPZ-4 score (P = .01), and 0.02 m/s slower time-updated gait speed (P < .0001). Age accentuated these effects. In the oldest age-quartile, higher citrate was associated with 1.64 higher odds of prevalent NCI, 0.17 SD lower NPZ-4, and 0.04 m/s slower gait speed (P ≤ .01 for each). Similar associations were apparent with succinate in the oldest age-quintile, but not with gait speed. In participants without NCI at entry, higher citrate predicted a faster rate of neurocognitive decline. CONCLUSIONS: Higher plasma citrate and succinate are associated with worse cross-sectional and longitudinal measures of neurocognitive function and gait speed that are age-dependent, supporting the importance of altered bioenergetic metabolism in the pathogenesis of NCI in older PWH.


Asunto(s)
Infecciones por VIH , Ácido Succínico , Adulto , Anciano , Ácido Cítrico , Estudios Transversales , Infecciones por VIH/complicaciones , Humanos , Persona de Mediana Edad , Estudios Prospectivos
4.
Anesth Analg ; 133(4): 924-932, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33591116

RESUMEN

BACKGROUND: Children with mitochondrial disease undergo anesthesia for a wide array of surgical procedures. However, multiple medications used for their perioperative care can affect mitochondrial function. Defects in function of the mitochondrial electron transport chain (ETC) can lead to a profound hypersensitivity to sevoflurane in children. We studied the sensitivities to sevoflurane, during mask induction and maintenance of general anesthesia, in children presenting for muscle biopsies for diagnosis of mitochondrial disease. METHODS: In this multicenter study, 91 children, aged 6 months to 16 years, presented to the operating room for diagnostic muscle biopsy for presumptive mitochondrial disease. General anesthesia was induced by a slow increase of inhaled sevoflurane concentration. The primary end point, end-tidal (ET) sevoflurane necessary to achieve a bispectral index (BIS) of 60, was recorded. Secondary end points were maximal sevoflurane used to maintain a BIS between 40 and 60 during the case, and maximum and minimum heart rate and blood pressures. After induction, general anesthesia was maintained according to the preferences of the providers directing the cases. Primary data were analyzed comparing data from patients with complex I deficiencies to other groups using nonparametric statistics in SPSS v.27. RESULTS: The median sevoflurane concentration to reach BIS of 60 during inductions (ET sevoflurane % [BIS = 60]) was significantly lower for patients with complex I defects (0.98%; 95% confidence interval [CI], 0.5-1.4) compared to complex II (1.95%; 95% CI, 1.2-2.7; P < .001), complex III (2.0%; 95% CI, 0.7-3.5; P < .001), complex IV (2.0%; 95% CI, 1.7-3.2; P < .001), and normal groups (2.2%; 95% CI, 1.8-3.0; P < .001). The sevoflurane sensitivities of complex I patients did not reach significance when compared to patients diagnosed with mitochondrial disease but without an identifiable ETC abnormality (P = .172). Correlation of complex I activity with ET sevoflurane % (BIS = 60) gave a Spearman's coefficient of 0.505 (P < .001). The differences in sensitivities between groups were less during the maintenance of the anesthetic than during induction. CONCLUSIONS: The data indicate that patients with complex I dysfunction are hypersensitive to sevoflurane compared to normal patients. Hypersensitivity was less common in patients presenting with other mitochondrial defects or without a mitochondrial diagnosis.


Asunto(s)
Anestesia General/efectos adversos , Anestésicos por Inhalación/efectos adversos , Hipersensibilidad a las Drogas/etiología , Complejo I de Transporte de Electrón/deficiencia , Enfermedades Mitocondriales/complicaciones , Músculo Esquelético/enzimología , Sevoflurano/efectos adversos , Adolescente , Factores de Edad , Anestésicos por Inhalación/administración & dosificación , Biopsia , Estudios de Casos y Controles , Niño , Preescolar , Hipersensibilidad a las Drogas/diagnóstico , Femenino , Humanos , Lactante , Masculino , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/enzimología , Músculo Esquelético/patología , Ohio , Medición de Riesgo , Factores de Riesgo , Sevoflurano/administración & dosificación , Resultado del Tratamiento , Washingtón
5.
J Lipid Res ; 61(12): 1707-1719, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32998976

RESUMEN

Phospholipids, including ether phospholipids, are composed of numerous isomeric and isobaric species that have the same backbone and acyl chains. This structural resemblance results in similar fragmentation patterns by collision-induced dissociation of phospholipids regardless of class, yielding complicated MS/MS spectra when isobaric species are analyzed together. Furthermore, the presence of isobaric species can lead to misassignment of species when made solely based on their molecular weights. In this study, we used normal-phase HPLC for ESI-MS/MS analysis of phospholipids from bovine heart mitochondria. Class separation by HPLC eliminates chances for misidentification of isobaric species from different classes of phospholipids. Chromatography yields simple MS/MS spectra without interference from isobaric species, allowing clear identification of peaks corresponding to fragmented ions containing monoacylglycerol backbone derived from losing one acyl chain. Using these fragmented ions, we characterized individual and isomeric species in each class of mitochondrial phospholipids, including unusual species, such as PS, containing an ether linkage and species containing odd-numbered acyl chains in cardiolipin, PS, PI, and PG. We also characterized monolysocardiolipin and dilysocardiolipin, the least abundant but nevertheless important mitochondrial phospholipids. The results clearly show the power of HPLC-MS/MS for identification and characterization of phospholipids, including minor species.


Asunto(s)
Cromatografía Líquida de Alta Presión , Mitocondrias Cardíacas/química , Fosfolípidos/análisis , Espectrometría de Masas en Tándem , Animales , Bovinos
6.
Hum Mol Genet ; 27(11): 1913-1926, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29566152

RESUMEN

Primary ovarian insufficiency (POI) is characterized by amenorrhea and loss or dysfunction of ovarian follicles prior to the age of 40. POI has been associated with autosomal recessive mutations in genes involving hormonal signaling and folliculogenesis, however, the genetic etiology of POI most often remains unknown. Here we report MRPS22 homozygous missense variants c.404G>A (p.R135Q) and c.605G>A (p.R202H) identified in four females from two independent consanguineous families as a novel genetic cause of POI in adolescents. Both missense mutations identified in MRPS22 are rare, occurred in highly evolutionarily conserved residues, and are predicted to be deleterious to protein function. In contrast to prior reports of mutations in MRPS22 associated with severe mitochondrial disease, the POI phenotype is far less severe. Consistent with this genotype-phenotype correlation, mitochondrial defects in oxidative phosphorylation or rRNA levels were not detected in fibroblasts derived from the POI patients, suggesting a non-bioenergetic or tissue-specific mitochondrial defect. Furthermore, we demonstrate in a Drosophila model that mRpS22 deficiency specifically in somatic cells of the ovary had no effect on fertility, whereas flies with mRpS22 deficiency specifically in germ cells were infertile and agametic, demonstrating a cell autonomous requirement for mRpS22 in germ cell development. These findings collectively identify that MRPS22, a component of the small mitochondrial ribosome subunit, is critical for ovarian development and may therefore provide insight into the pathophysiology and treatment of ovarian dysfunction.


Asunto(s)
Proteínas de Drosophila/genética , Fertilidad/genética , Proteínas Mitocondriales/genética , Insuficiencia Ovárica Primaria/genética , Proteínas Ribosómicas/genética , Adolescente , Adulto , Amenorrea/genética , Amenorrea/patología , Animales , Modelos Animales de Enfermedad , Drosophila/genética , Femenino , Fertilidad/fisiología , Homocigoto , Humanos , Menopausia Prematura/genética , Mutación Missense/genética , Folículo Ovárico/patología , Insuficiencia Ovárica Primaria/patología , Adulto Joven
7.
Annu Rev Pharmacol Toxicol ; 57: 535-565, 2017 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-27860548

RESUMEN

Mitochondria have emerged as key participants in and regulators of myocardial injury during ischemia and reperfusion. This review examines the sites of damage to cardiac mitochondria during ischemia and focuses on the impact of these defects. The concept that mitochondrial damage during ischemia leads to cardiac injury during reperfusion is addressed. The mechanisms that translate ischemic mitochondrial injury into cellular damage, during both ischemia and early reperfusion, are examined. Next, we discuss strategies that modulate and counteract these mechanisms of mitochondrial-driven injury. The new concept that mitochondria are not merely stochastic sites of oxidative and calcium-mediated injury but that they activate cellular responses of mitochondrial remodeling and cellular reactions that modulate the balance between cell death and recovery is reviewed, and the therapeutic implications of this concept are discussed.


Asunto(s)
Fármacos Cardiovasculares/uso terapéutico , Precondicionamiento Isquémico Miocárdico/métodos , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Animales , Fármacos Cardiovasculares/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/fisiología , Humanos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/patología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/patología
8.
Am J Physiol Endocrinol Metab ; 317(6): E999-E1014, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31526287

RESUMEN

Insulin resistance has wide-ranging effects on metabolism, but there are knowledge gaps regarding the tissue origins of systemic metabolite patterns and how patterns are altered by fitness and metabolic health. To address these questions, plasma metabolite patterns were determined every 5 min during exercise (30 min, ∼45% of V̇o2peak, ∼63 W) and recovery in overnight-fasted sedentary, obese, insulin-resistant women under controlled conditions of diet and physical activity. We hypothesized that improved fitness and insulin sensitivity following a ∼14-wk training and weight loss intervention would lead to fixed workload plasma metabolomics signatures reflective of metabolic health and muscle metabolism. Pattern analysis over the first 15 min of exercise, regardless of pre- versus postintervention status, highlighted anticipated increases in fatty acid tissue uptake and oxidation (e.g., reduced long-chain fatty acids), diminution of nonoxidative fates of glucose [e.g., lowered sorbitol-pathway metabolites and glycerol-3-galactoside (possible glycerolipid synthesis metabolite)], and enhanced tissue amino acid use (e.g., drops in amino acids; modest increase in urea). A novel observation was that exercise significantly increased several xenometabolites ("non-self" molecules, from microbes or foods), including benzoic acid-salicylic acid-salicylaldehyde, hexadecanol-octadecanol-dodecanol, and chlorogenic acid. In addition, many nonannotated metabolites changed with exercise. Although exercise itself strongly impacted the global metabolome, there were surprisingly few intervention-associated differences despite marked improvements in insulin sensitivity, fitness, and adiposity. These results and previously reported plasma acylcarnitine profiles support the principle that most metabolic changes during submaximal aerobic exercise are closely tethered to absolute ATP turnover rate (workload), regardless of fitness or metabolic health status.


Asunto(s)
Aminoácidos/metabolismo , Ejercicio Físico/fisiología , Ácidos Grasos/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina , Metaboloma , Obesidad/terapia , Conducta Sedentaria , Programas de Reducción de Peso , Adiposidad , Adulto , Ayuno , Femenino , Humanos , Metabolómica , Persona de Mediana Edad , Obesidad/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Aptitud Física
9.
Circ Res ; 118(10): 1593-611, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27174952

RESUMEN

Altered mitochondrial metabolism is the underlying basis for the increased sensitivity in the aged heart to stress. The aged heart exhibits impaired metabolic flexibility, with a decreased capacity to oxidize fatty acids and enhanced dependence on glucose metabolism. Aging impairs mitochondrial oxidative phosphorylation, with a greater role played by the mitochondria located between the myofibrils, the interfibrillar mitochondria. With aging, there is a decrease in activity of complexes III and IV, which account for the decrease in respiration. Furthermore, aging decreases mitochondrial content among the myofibrils. The end result is that in the interfibrillar area, there is ≈50% decrease in mitochondrial function, affecting all substrates. The defective mitochondria persist in the aged heart, leading to enhanced oxidant production and oxidative injury and the activation of oxidant signaling for cell death. Aging defects in mitochondria represent new therapeutic targets, whether by manipulation of the mitochondrial proteome, modulation of electron transport, activation of biogenesis or mitophagy, or the regulation of mitochondrial fission and fusion. These mechanisms provide new ways to attenuate cardiac disease in elders by preemptive treatment of age-related defects, in contrast to the treatment of disease-induced dysfunction.


Asunto(s)
Envejecimiento/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Corazón/crecimiento & desarrollo , Mitocondrias Cardíacas/metabolismo , Biogénesis de Organelos , Envejecimiento/patología , Animales , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Corazón/fisiopatología , Humanos , Estrés Oxidativo
10.
Circulation ; 133(8): 698-705, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26819376

RESUMEN

BACKGROUND: Significant evidence indicates that the failing heart is energy starved. During the development of heart failure, the capacity of the heart to utilize fatty acids, the chief fuel, is diminished. Identification of alternate pathways for myocardial fuel oxidation could unveil novel strategies to treat heart failure. METHODS AND RESULTS: Quantitative mitochondrial proteomics was used to identify energy metabolic derangements that occur during the development of cardiac hypertrophy and heart failure in well-defined mouse models. As expected, the amounts of proteins involved in fatty acid utilization were downregulated in myocardial samples from the failing heart. Conversely, expression of ß-hydroxybutyrate dehydrogenase 1, a key enzyme in the ketone oxidation pathway, was increased in the heart failure samples. Studies of relative oxidation in an isolated heart preparation using ex vivo nuclear magnetic resonance combined with targeted quantitative myocardial metabolomic profiling using mass spectrometry revealed that the hypertrophied and failing heart shifts to oxidizing ketone bodies as a fuel source in the context of reduced capacity to oxidize fatty acids. Distinct myocardial metabolomic signatures of ketone oxidation were identified. CONCLUSIONS: These results indicate that the hypertrophied and failing heart shifts to ketone bodies as a significant fuel source for oxidative ATP production. Specific metabolite biosignatures of in vivo cardiac ketone utilization were identified. Future studies aimed at determining whether this fuel shift is adaptive or maladaptive could unveil new therapeutic strategies for heart failure.


Asunto(s)
Dieta Cetogénica/métodos , Ácidos Grasos/metabolismo , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Cuerpos Cetónicos/metabolismo , Animales , Femenino , Perfilación de la Expresión Génica/métodos , Insuficiencia Cardíaca/dietoterapia , Ratones , Ratones Endogámicos C57BL
11.
Am J Physiol Endocrinol Metab ; 313(6): E690-E698, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28830869

RESUMEN

The period around bariatric surgery offers a unique opportunity to characterize metabolism responses to dynamic shifts in energy, gut function, and anesthesia. We analyzed plasma acylcarnitines in obese women (n = 17) sampled in the overnight fasted/postabsorptive state approximately 1-2 wk before surgery (condition A), the morning of surgery (prior restriction to a 48-h clear liquid diet coupled in some cases a standard polyethylene glycol gut evacuation: condition B), and following induction of anesthesia (condition C). Comparisons tested if 1) plasma acylcarnitine derivatives reflective of fatty acid oxidation (FAO) and xenometabolism would be significantly increased and decreased, respectively, by preoperative gut preparation/negative energy balance (condition A vs. B), and 2) anesthesia would acutely depress markers of FAO. Acylcarnitines associated with fat mobilization and FAO were significantly increased in condition B: long-chain acylcarnitines (i.e., C18:1, ~70%), metabolites from active but incomplete FAO [i.e., C14:1 (161%) and C14:2 (102%)] and medium- to short-chain acylcarnitines [i.e., C2 (91%), R-3-hydroxybutyryl-(245%), C6 (45%), and cis-3,4-methylene-heptanoyl-(17%), etc.]. Branched-chain amino acid markers displayed disparate patterns [i.e., isobutyryl-(40% decreased) vs. isovaleryl carnitine (51% increased)]. Anesthesia reduced virtually every acylcarnitine. These results are consistent with a fasting-type metabolic phenotype coincident with the presurgical "gut preparation" phase of bariatric surgery, and a major and rapid alteration of both fat and amino acid metabolism with onset of anesthesia. Whether presurgical or anesthesia-associated metabolic shifts in carnitine and fuel metabolism impact patient outcomes or surgical risks remains to be evaluated experimentally.


Asunto(s)
Aminoácidos de Cadena Ramificada/metabolismo , Anestesia , Cirugía Bariátrica , Carnitina/análogos & derivados , Catárticos/efectos adversos , Metabolismo de los Lípidos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Adulto , Anestesia/efectos adversos , Anestesia/métodos , Cirugía Bariátrica/efectos adversos , Cirugía Bariátrica/métodos , Carnitina/sangre , Catárticos/farmacología , Ayuno/metabolismo , Ácidos Grasos/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Obesidad Mórbida/metabolismo , Obesidad Mórbida/cirugía , Oxidación-Reducción/efectos de los fármacos , Cuidados Preoperatorios/efectos adversos , Cuidados Preoperatorios/métodos , Adulto Joven
12.
Mol Genet Metab ; 120(4): 363-369, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28190699

RESUMEN

While selectively quantifying acylcarnitines in thousands of patient samples using UHPLC-MS/MS, we have occasionally observed unidentified branched-chain C8 acylcarnitines. Such observations are not possible using tandem MS methods, which generate pseudo-quantitative acylcarnitine "profiles". Since these "profiles" select for mass alone, they cannot distinguish authentic signal from isobaric and isomeric interferences. For example, some of the samples containing branched-chain C8 acylcarnitines were, in fact, expanded newborn screening false positive "profiles" for medium-chain acyl-CoA dehydrogenase deficiency (MCADD). Using our fast, highly selective, and quantitatively accurate UHPLC-MS/MS acylcarnitine determination method, we corrected the false positive tandem MS results and reported the sample results as normal for octanoylcarnitine (the marker for MCADD). From instances such as these, we decided to further investigate the presence of branched-chain C8 acylcarnitines in patient samples. To accomplish this, we synthesized and chromatographically characterized several branched-chain C8 acylcarnitines (in addition to valproylcarnitine): 2-methylheptanoylcarnitine, 6-methylheptanoylcarnitine, 2,2-dimethylhexanoylcarnitine, 3,3-dimethylhexanoylcarnitine, 3,5-dimethylhexanoylcarnitine, 2-ethylhexanoylcarnitine, and 2,4,4-trimethylpentanoylcarnitine. We then compared their behavior with branched-chain C8 acylcarnitines observed in patient samples and demonstrated our ability to chromographically resolve, and thus distinguish, octanoylcarnitine from branched-chain C8 acylcarnitines, correcting false positive MCADD results from expanded newborn screening.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Carnitina/análogos & derivados , Carnitina/metabolismo , Errores Innatos del Metabolismo Lipídico/diagnóstico , Tamizaje Neonatal/normas , Carnitina/síntesis química , Carnitina/aislamiento & purificación , Cromatografía Líquida de Alta Presión/métodos , Reacciones Falso Positivas , Humanos , Recién Nacido , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem
13.
Mol Genet Metab ; 120(4): 342-349, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28202214

RESUMEN

Mutations in ECHS1 result in short-chain enoyl-CoA hydratase (SCEH) deficiency which mainly affects the catabolism of various amino acids, particularly valine. We describe a case compound heterozygous for ECHS1 mutations c.836T>C (novel) and c.8C>A identified by whole exome sequencing of proband and parents. SCEH deficiency was confirmed with very low SCEH activity in fibroblasts and nearly absent immunoreactivity of SCEH. The patient had a severe neonatal course with elevated blood and cerebrospinal fluid lactate and pyruvate concentrations, high plasma alanine and slightly low plasma cystine. 2-Methyl-2,3-dihydroxybutyric acid was markedly elevated as were metabolites of the three branched-chain α-ketoacids on urine organic acids analysis. These urine metabolites notably decreased when lactic acidosis decreased in blood. Lymphocyte pyruvate dehydrogenase complex (PDC) activity was deficient, but PDC and α-ketoglutarate dehydrogenase complex activities in cultured fibroblasts were normal. Oxidative phosphorylation analysis on intact digitonin-permeabilized fibroblasts was suggestive of slightly reduced PDC activity relative to control range in mitochondria. We reviewed 16 other cases with mutations in ECHS1 where PDC activity was also assayed in order to determine how common and generalized secondary PDC deficiency is associated with primary SCEH deficiency. For reasons that remain unexplained, we find that about half of cases with primary SCEH deficiency also exhibit secondary PDC deficiency. The patient died on day-of-life 39, prior to establishing his diagnosis, highlighting the importance of early and rapid neonatal diagnosis because of possible adverse effects of certain therapeutic interventions, such as administration of ketogenic diet, in this disorder. There is a need for better understanding of the pathogenic mechanisms and phenotypic variability in this relatively recently discovered disorder.


Asunto(s)
Enoil-CoA Hidratasa/deficiencia , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/mortalidad , Análisis de Secuencia de ADN/métodos , Enoil-CoA Hidratasa/genética , Exoma , Humanos , Recién Nacido , Masculino , Polimorfismo de Nucleótido Simple , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/genética
14.
Mol Genet Metab ; 120(3): 213-222, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27913098

RESUMEN

Mutations in SUCLA2 result in succinyl-CoA ligase (ATP-forming) or succinyl-CoA synthetase (ADP-forming) (A-SCS) deficiency, a mitochondrial tricarboxylic acid cycle disorder. The phenotype associated with this gene defect is largely encephalomyopathy. We describe two siblings compound heterozygous for SUCLA2 mutations, c.985A>G (p.M329V) and c.920C>T (p.A307V), with parents confirmed as carriers of each mutation. We developed a new LC-MS/MS based enzyme assay to demonstrate the decreased SCS activity in the siblings with this unique genotype. Both siblings shared bilateral progressive hearing loss, encephalopathy, global developmental delay, generalized myopathy, and dystonia with choreoathetosis. Prior to diagnosis and because of lactic acidosis and low activity of muscle pyruvate dehydrogenase complex (PDC), sibling 1 (S1) was placed on dichloroacetate, while sibling 2 (S2) was on a ketogenic diet. S1 developed severe cyclic vomiting refractory to therapy, while S2 developed Leigh syndrome, severe GI dysmotility, intermittent anemia, hypogammaglobulinemia and eventually succumbed to his disorder. The mitochondrial DNA contents in skeletal muscle (SM) were normal in both siblings. Pyruvate dehydrogenase complex, ketoglutarate dehydrogenase complex, and several mitochondrial electron transport chain (ETC) activities were low or at the low end of the reference range in frozen SM from S1 and/or S2. In contrast, activities of PDC, other mitochondrial enzymes of pyruvate metabolism, ETC and, integrated oxidative phosphorylation, in skin fibroblasts were not significantly impaired. Although we show that propionyl-CoA inhibits PDC, it does not appear to account for decreased PDC activity in SM. A better understanding of the mechanisms of phenotypic variability and the etiology for tissue-specific secondary deficiencies of mitochondrial enzymes of oxidative metabolism, and independently mitochondrial DNA depletion (common in other cases of A-SCS deficiency), is needed given the implications for control of lactic acidosis and possible clinical management.


Asunto(s)
Enfermedades Mitocondriales/genética , Músculo Esquelético/enzimología , Polimorfismo de Nucleótido Simple , Succinato-CoA Ligasas/deficiencia , Adolescente , Niño , ADN Mitocondrial/genética , Resultado Fatal , Humanos , Masculino , Enfermedades Mitocondriales/enzimología , Músculo Esquelético/metabolismo , Eliminación de Secuencia , Hermanos , Succinato-CoA Ligasas/genética
15.
Exp Physiol ; 102(1): 48-69, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27730694

RESUMEN

NEW FINDINGS: What is the central question of this study? Does improved metabolic health and insulin sensitivity following a weight-loss and fitness intervention in sedentary, obese women alter exercise-associated fuel metabolism and incomplete mitochondrial fatty acid oxidation (FAO), as tracked by blood acylcarnitine patterns? What is the main finding and its importance? Despite improved fitness and blood sugar control, indices of incomplete mitochondrial FAO increased in a similar manner in response to a fixed load acute exercise bout; this indicates that intramitochondrial muscle FAO is inherently inefficient and is tethered directly to ATP turnover. With insulin resistance or type 2 diabetes mellitus, mismatches between mitochondrial fatty acid fuel delivery and oxidative phosphorylation/tricarboxylic acid cycle activity may contribute to inordinate accumulation of short- or medium-chain acylcarnitine fatty acid derivatives [markers of incomplete long-chain fatty acid oxidation (FAO)]. We reasoned that incomplete FAO in muscle would be ameliorated concurrent with improved insulin sensitivity and fitness following a ∼14 week training and weight-loss intervention in obese, sedentary, insulin-resistant women. Contrary to this hypothesis, overnight-fasted and exercise-induced plasma C4-C14 acylcarnitines did not differ between pre- and postintervention phases. These metabolites all increased robustly with exercise (∼45% of pre-intervention peak oxygen consumption) and decreased during a 20 min cool-down. This supports the idea that, regardless of insulin sensitivity and fitness, intramitochondrial muscle ß-oxidation and attendant incomplete FAO are closely tethered to absolute ATP turnover rate. Acute exercise also led to branched-chain amino acid acylcarnitine derivative patterns suggestive of rapid and transient diminution of branched-chain amino acid flux through the mitochondrial branched-chain ketoacid dehydrogenase complex. We confirmed our prior novel observation that a weight-loss/fitness intervention alters plasma xenometabolites [i.e. cis-3,4-methylene-heptanoylcarnitine and γ-butyrobetaine (a co-metabolite possibly derived in part from gut bacteria)], suggesting that host metabolic health regulated gut microbe metabolism. Finally, we considered whether acylcarnitine metabolites signal to muscle-innervating afferents; palmitoylcarnitine at concentrations as low as 1-10 µm activated a subset (∼2.5-5%) of these neurons ex vivo. This supports the hypothesis that in addition to tracking exercise-associated shifts in fuel metabolism, muscle acylcarnitines act as signals of exertion to short-loop somatosensory-motor circuits or to the brain.


Asunto(s)
Biomarcadores/metabolismo , Carnitina/análogos & derivados , Ejercicio Físico/fisiología , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Neuronas Aferentes/metabolismo , Neuronas Aferentes/fisiología , Adenosina Trifosfato/metabolismo , Adulto , Aminoácidos de Cadena Ramificada/metabolismo , Carnitina/metabolismo , Ciclo del Ácido Cítrico/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Ácidos Grasos/metabolismo , Femenino , Humanos , Resistencia a la Insulina/fisiología , Persona de Mediana Edad , Mitocondrias Musculares/metabolismo , Músculo Esquelético/fisiopatología , Obesidad/metabolismo , Obesidad/fisiopatología , Oxidación-Reducción , Fosforilación Oxidativa , Consumo de Oxígeno/fisiología , Pérdida de Peso/fisiología
16.
Adv Exp Med Biol ; 982: 451-464, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28551802

RESUMEN

Mitochondria are the prime source of ATP in cardiomyocytes. Impairment of mitochondrial metabolism results in damage to existing proteins and DNA. Such deleterious effects are part and parcel of the aging process, reducing the ability of cardiomyocytes to counter stress, such as myocardial infarction and consequent reperfusion. In such conditions, mitochondria in the heart of aged individuals exhibit decreased oxidative phosphorylation, decreased ATP production, and increased net reactive oxygen species production; all of these effects are independent of the decrease in number of mitochondria that occurs in these situations. Rather than being associated with the mitochondrial population in toto, these defects are almost exclusively confined to those organelles positioned between myofibrils (interfibrillar mitochondria). It is in complex III and IV where these dysfunctional aspects are manifested. In an apparent effort to correct mitochondrial metabolic defects, affected organelles are to some extent eliminated by mitophagy; at the same time, new, unaffected organelles are generated by fission of mitochondria. Because these cardiac health issues are localized to specific mitochondria, these organelles offer potential targets for therapeutic approaches that could favorably affect the aging process in heart.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Cardiovasculares/metabolismo , Senescencia Celular , Metabolismo Energético , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Edad , Envejecimiento/patología , Animales , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Enfermedades Cardiovasculares/prevención & control , Senescencia Celular/efectos de los fármacos , Ingestión de Energía , Metabolismo Energético/efectos de los fármacos , Humanos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/ultraestructura , Dinámicas Mitocondriales , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , Conducta de Reducción del Riesgo
17.
Mol Genet Metab ; 119(1-2): 50-6, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27397597

RESUMEN

Glutaric aciduria type I (GA-I) is an autosomal recessive organic aciduria resulting from a functional deficiency of glutaryl-CoA dehydrogenase, encoded by GCDH. Two clinically indistinguishable diagnostic subgroups of GA-I are known; low and high excretors (LEs and HEs, respectively). Early medical and dietary interventions can result in significantly better outcomes and improved quality of life for patients with GA-I. We report on nine cases of GA-I LE patients all sharing the M405V allele with two cases missed by newborn screening (NBS) using tandem mass spectrometry (MS/MS). We describe a novel case with the known pathogenic M405V variant and a novel V133L variant, and present updated and previously unreported clinical, biochemical, functional and molecular data on eight other patients all sharing the M405V allele. Three of the nine patients are of African American ancestry, with two as siblings. GCDH activity was assayed in six of the nine patients and varied from 4 to 25% of the control mean. We support the use of urine glutarylcarnitine as a biochemical marker of GA-I by demonstrating that glutarylcarnitine is efficiently cleared by the kidney (50-90%) and that plasma and urine glutarylcarnitine follow a linear relationship. We report the allele frequencies for three known GA-I LE GCDH variants (M405V, V400M and R227P) and note that both the M405V and V400M variants are significantly more common in the population of African ancestry compared to the general population. This report highlights the M405V allele as another important molecular marker in patients with the GA-I LE phenotype. Therefore, the incorporation into newborn screening of molecular screening for the M405V and V400M variants in conjunction with MS/MS could help identify asymptomatic at-risk GA-I LE patients that could potentially be missed by current NBS programs.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Biomarcadores , Encefalopatías Metabólicas/genética , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Tamizaje Neonatal , Negro o Afroamericano/genética , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/fisiopatología , Femenino , Frecuencia de los Genes , Glutaratos/metabolismo , Humanos , Recién Nacido , Masculino , Mutación , Fenotipo , Espectrometría de Masas en Tándem
18.
J Biol Chem ; 289(14): 10223-34, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24558043

RESUMEN

Rat hearts were perfused with [1,2,3,4-(13)C4]palmitic acid (M+4), and the isotopic patterns of myocardial acylcarnitines and acyl-CoAs were analyzed using ultra-HPLC-MS/MS. The 91.2% (13)C enrichment in palmitoylcarnitine shows that little endogenous (M+0) palmitate contributed to its formation. The presence of M+2 myristoylcarnitine (95.7%) and M+2 acetylcarnitine (19.4%) is evidence for ß-oxidation of perfused M+4 palmitic acid. Identical enrichment data were obtained in the respective acyl-CoAs. The relative (13)C enrichment in M+4 (84.7%, 69.9%) and M+6 (16.2%, 17.8%) stearoyl- and arachidylcarnitine, respectively, clearly shows that the perfused palmitate is chain-elongated. The observed enrichment of (13)C in acetylcarnitine (19%), M+6 stearoylcarnitine (16.2%), and M+6 arachidylcarnitine (17.8%) suggests that the majority of two-carbon units for chain elongation are derived from ß-oxidation of [1,2,3,4-(13)C4]palmitic acid. These data are explained by conversion of the M+2 acetyl-CoA to M+2 malonyl-CoA, which serves as the acceptor for M+4 palmitoyl-CoA in chain elongation. Indeed, the (13)C enrichment in mitochondrial acetyl-CoA (18.9%) and malonyl-CoA (19.9%) are identical. No (13)C enrichment was found in acylcarnitine species with carbon chain lengths between 4 and 12, arguing against the simple reversal of fatty acid ß-oxidation. Furthermore, isolated, intact rat heart mitochondria 1) synthesize malonyl-CoA with simultaneous inhibition of carnitine palmitoyltransferase 1b and 2) catalyze the palmitoyl-CoA-dependent incorporation of (14)C from [2-(14)C]malonyl-CoA into lipid-soluble products. In conclusion, rat heart has the capability to chain-elongate fatty acids using mitochondria-derived two-carbon chain extenders. The data suggest that the chain elongation process is localized on the outer surface of the mitochondrial outer membrane.


Asunto(s)
Acetilcoenzima A/metabolismo , Inhibidores Enzimáticos/farmacología , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Ácido Palmítico/metabolismo , Ácido Palmítico/farmacología , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Inhibidores Enzimáticos/metabolismo , Malonil Coenzima A/metabolismo , Proteínas Musculares/metabolismo , Oxidación-Reducción , Palmitoil Coenzima A/metabolismo , Perfusión , Ratas , Ratas Endogámicas F344
19.
J Biol Chem ; 289(1): 251-63, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24265312

RESUMEN

CREB-binding protein (CBP)/p300 interacting transactivator with glutamic acid (Glu) and aspartic acid (Asp)-tail 2 (Cited2) was recently shown to be essential for gluconeogenesis in the adult mouse. The metabolic function of Cited2 in mouse embryonic stem cells (mESCs) remains elusive. In the current study, the metabolism of glucose was investigated in mESCs, which contained a deletion in the gene for Cited2 (Cited2(Δ/-)). Compared with its parental wild type counterpart, Cited2(Δ/-) ESCs have enhanced glycolysis, alternations in mitochondria morphology, reduced glucose oxidation, and decreased ATP content. Cited2 is recruited to the hexokinase 1 (HK1) gene promoter to regulate transcription of HK1, which coordinates glucose metabolism in wild type ESCs. Reduced glucose oxidation and enhanced glycolytic activity in Cited2(Δ/-) ESCs correlates with defective differentiation during hypoxia, which is reflected in an increased expression of pluripotency marker (Oct4) and epiblast marker (Fgf5) and decreased expression of lineage specification markers (T, Gata-6, and Cdx2). Knockdown of hypoxia inducible factor-1α in Cited2(Δ/-) ESCs re-initiates the expression of differentiation markers T and Gata-6. Taken together, a deletion of Cited2 in mESCs results in abnormal mitochondrial morphology and impaired glucose metabolism, which correlates with a defective cell fate decision.


Asunto(s)
Células Madre Embrionarias/metabolismo , Glucólisis/fisiología , Mitocondrias/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Transcripción Genética/fisiología , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/genética , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Hipoxia de la Célula/fisiología , Células Madre Embrionarias/citología , Glucosa/genética , Glucosa/metabolismo , Hexoquinasa/biosíntesis , Hexoquinasa/genética , Ratones , Ratones Noqueados , Mitocondrias/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oxidación-Reducción , Proteínas Represoras/genética , Transactivadores/genética
20.
J Biol Chem ; 289(9): 5914-24, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24407292

RESUMEN

The mammalian heart, the body's largest energy consumer, has evolved robust mechanisms to tightly couple fuel supply with energy demand across a wide range of physiologic and pathophysiologic states, yet, when compared with other organs, relatively little is known about the molecular machinery that directly governs metabolic plasticity in the heart. Although previous studies have defined Kruppel-like factor 15 (KLF15) as a transcriptional repressor of pathologic cardiac hypertrophy, a direct role for the KLF family in cardiac metabolism has not been previously established. We show in human heart samples that KLF15 is induced after birth and reduced in heart failure, a myocardial expression pattern that parallels reliance on lipid oxidation. Isolated working heart studies and unbiased transcriptomic profiling in Klf15-deficient hearts demonstrate that KLF15 is an essential regulator of lipid flux and metabolic homeostasis in the adult myocardium. An important mechanism by which KLF15 regulates its direct transcriptional targets is via interaction with p300 and recruitment of this critical co-activator to promoters. This study establishes KLF15 as a key regulator of myocardial lipid utilization and is the first to implicate the KLF transcription factor family in cardiac metabolism.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Metabolismo de los Lípidos , Proteínas Musculares/metabolismo , Miocardio/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patología , Línea Celular , Proteínas de Unión al ADN/genética , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Miocardio/patología , Proteínas Nucleares/genética , Oxidación-Reducción , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA