Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-36356723

RESUMEN

Recent trials have reported the ability of triheptanoin to improve clinical outcomes for the severe symptoms associated with long-chain fatty acid oxidation disorders, including very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency. However, the milder myopathic symptoms are still challenging to treat satisfactorily. Myopathic pathogenesis is multifactorial, but oxidative stress is an important component. We have previously shown that metabolic stress increases the oxidative burden in VLCAD-deficient cell lines and can deplete the antioxidant glutathione (GSH). We investigated whether medium-chain fatty acids provide protection against GSH depletion during metabolic stress in VLCAD-deficient fibroblasts. To investigate the effect of differences in anaplerotic capacity, we included both even-(octanoate) and odd-numbered (heptanoate) medium-chain fatty acids. Overall, we show that modulation of the concentration of medium-chain fatty acids in culture media affects levels of GSH retained during metabolic stress in VLCAD-deficient cell lines but not in controls. Lowered glutamine concentration in the culture media during metabolic stress led to GSH depletion and decreased viability in VLCAD deficient cells, which could be rescued by both heptanoate and octanoate in a dose-dependent manner. Unlike GSH levels, the levels of total thiols increased after metabolic stress exposure, the size of this increase was not affected by differences in cell culture medium concentrations of glutamine, heptanoate or octanoate. Addition of a PPAR agonist further exacerbated stress-related GSH-depletion and viability loss, requiring higher concentrations of fatty acids to restore GSH levels and cell viability. Both odd- and even-numbered medium-chain fatty acids efficiently protect VLCADdeficient cells against metabolic stress-induced antioxidant depletion.


Asunto(s)
Acil-CoA Deshidrogenasa de Cadena Larga , Caprilatos , Caprilatos/metabolismo , Acil-CoA Deshidrogenasa de Cadena Larga/metabolismo , Heptanoatos/metabolismo , Antioxidantes , Glutamina , Ácidos Grasos/metabolismo , Glutatión , Medios de Cultivo
2.
Free Radic Res ; 56(7-8): 511-525, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36480241

RESUMEN

Flavin adenine dinucleotide (FAD) synthase (EC 2.7.7.2), encoded by human flavin adenine dinucleotide synthetase 1 (FLAD1), catalyzes the last step of the pathway converting riboflavin (Rf) into FAD. FLAD1 variations were identified as a cause of LSMFLAD (lipid storage myopathy due to FAD synthase deficiency, OMIM #255100), resembling Multiple Acyl-CoA Dehydrogenase Deficiency, sometimes treatable with high doses of Rf; no alternative therapeutic strategies are available. We describe here cell morphological and mitochondrial alterations in dermal fibroblasts derived from a LSMFLAD patient carrying a homozygous truncating FLAD1 variant (c.745C > T) in exon 2. Despite a severe decrease in FAD synthesis rate, the patient had decreased cellular levels of Rf and flavin mononucleotide and responded to Rf treatment. We hypothesized that disturbed flavin homeostasis and Rf-responsiveness could be due to a secondary impairment in the expression of the Rf transporter 2 (RFVT2), encoded by SLC52A2, in the frame of an adaptive retrograde signaling to mitochondrial dysfunction. Interestingly, an antioxidant response element (ARE) is found in the region upstream of the transcriptional start site of SLC52A2. Accordingly, we found that abnormal mitochondrial morphology and impairments in bioenergetics were accompanied by increased cellular reactive oxygen species content and mtDNA oxidative damage. Concomitantly, an active response to mitochondrial stress is suggested by increased levels of PPARγ-co-activator-1α and Peroxiredoxin III. In this scenario, the treatment with high doses of Rf might compensate for the secondary RFVT2 molecular defect, providing a molecular rationale for the Rf responsiveness in patients with loss of function variants in FLAD1 exon 2.HIGHLIGHTSFAD synthase deficiency alters mitochondrial morphology and bioenergetics;FAD synthase deficiency triggers a mitochondrial retrograde response;FAD synthase deficiency evokes nuclear signals that adapt the expression of RFVT2.


Asunto(s)
Flavina-Adenina Dinucleótido , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa , Humanos , Flavina-Adenina Dinucleótido/genética , Flavina-Adenina Dinucleótido/metabolismo , Flavina-Adenina Dinucleótido/uso terapéutico , Riboflavina/genética , Riboflavina/metabolismo , Riboflavina/uso terapéutico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Exones , Mononucleótido de Flavina/genética , Mononucleótido de Flavina/uso terapéutico
3.
Biomolecules ; 11(7)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209852

RESUMEN

Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a heterogeneous, debilitating, and complex disease. Along with disabling fatigue, ME/CFS presents an array of other core symptoms, including autonomic nervous system (ANS) dysfunction, sustained inflammation, altered energy metabolism, and mitochondrial dysfunction. Here, we evaluated patients' symptomatology and the mitochondrial metabolic parameters in peripheral blood mononuclear cells (PBMCs) and plasma from a clinically well-characterised cohort of six ME/CFS patients compared to age- and gender-matched controls. We performed a comprehensive cellular assessment using bioenergetics (extracellular flux analysis) and protein profiles (quantitative mass spectrometry-based proteomics) together with self-reported symptom measures of fatigue, ANS dysfunction, and overall physical and mental well-being. This ME/CFS cohort presented with severe fatigue, which correlated with the severity of ANS dysfunction and overall physical well-being. PBMCs from ME/CFS patients showed significantly lower mitochondrial coupling efficiency. They exhibited proteome alterations, including altered mitochondrial metabolism, centred on pyruvate dehydrogenase and coenzyme A metabolism, leading to a decreased capacity to provide adequate intracellular ATP levels. Overall, these results indicate that PBMCs from ME/CFS patients have a decreased ability to fulfill their cellular energy demands.


Asunto(s)
Síndrome de Fatiga Crónica/sangre , Síndrome de Fatiga Crónica/inmunología , Síndrome de Fatiga Crónica/fisiopatología , Adulto , Células Sanguíneas/citología , Estudios de Cohortes , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Femenino , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Humanos , Leucocitos Mononucleares/citología , Persona de Mediana Edad , Mitocondrias/metabolismo , Proyectos Piloto , Proteoma/metabolismo , Proteómica/métodos
4.
J Inherit Metab Dis ; 44(5): 1215-1225, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33973257

RESUMEN

Ethylmalonic acid (EMA) is a major and potentially cytotoxic metabolite associated with short-chain acyl-CoA dehydrogenase (SCAD) deficiency, a condition whose status as a disease is uncertain. Unexplained high EMA is observed in some individuals with complex neurological symptoms, who carry the SCAD gene (ACADS) variants, c.625G>A and c.511C>T. The variants have a high allele frequency in the general population, but are significantly overrepresented in individuals with elevated EMA. This has led to the idea that these variants need to be associated with variants in other genes to cause hyperexcretion of ethylmalonic acid and possibly a diseased state. Ethylmalonyl-CoA decarboxylase (ECHDC1) has been described and characterized as an EMA metabolite repair enzyme, however, its clinical relevance has never been investigated. In this study, we sequenced the ECHDC1 gene (ECHDC1) in 82 individuals, who were reported with unexplained high EMA levels due to the presence of the common ACADS variants only. Three individuals with ACADS c.625G>A variants were found to be heterozygous for ECHDC1 loss-of-function variants. Knockdown experiments of ECHDC1, in healthy human cells with different ACADS c.625G>A genotypes, showed that ECHDC1 haploinsufficiency and homozygosity for the ACADS c.625G>A variant had a synergistic effect on cellular EMA excretion. This study reports the first cases of ECHDC1 gene defects in humans and suggests that ECHDC1 may be involved in elevated EMA excretion in only a small group of individuals with the common ACADS variants. However, a direct link between ECHDC1/ACADS deficiency, EMA and disease could not be proven.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Variación Genética , Errores Innatos del Metabolismo Lipídico/genética , Malonatos/metabolismo , Enzima Bifuncional Peroxisomal/genética , Acil-CoA Deshidrogenasa/genética , Alelos , Frecuencia de los Genes , Genotipo , Células HEK293 , Humanos , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(6): 166100, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33549744

RESUMEN

Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is the most common inborn long-chain fatty acid oxidation (FAO) disorder. VLCAD deficiency is characterized by distinct phenotypes. The severe phenotypes are potentially life-threatening and affect the heart or liver, with a comparatively milder phenotype characterized by myopathic symptoms. There is an unmet clinical need for effective treatment options for the myopathic phenotype. The molecular mechanisms driving the gradual decrease in mitochondrial function and associated alterations of muscle fibers are unclear. The peroxisome proliferator-activated receptor (PPAR) pan-agonist bezafibrate is a potent modulator of FAO and multiple other mitochondrial functions and has been proposed as a potential medication for myopathic cases of long-chain FAO disorders. In vitro experiments have demonstrated the ability of bezafibrate to increase VLCAD expression and activity. However, the outcome of small-scale clinical trials has been controversial. We found VLCAD deficient patient fibroblasts to have an increased oxidative stress burden and deranged mitochondrial bioenergetic capacity, compared to controls. Applying heat stress under fasting conditions to bezafibrate pretreated patient cells, caused a marked further increase of mitochondrial superoxide levels. Patient cells failed to maintain levels of the essential thiol peptide antioxidant glutathione and experienced a decrease in cellular viability. Our findings indicate that chronic PPAR activation is a plausible initiator of long-term pathogenesis in VLCAD deficiency. Our findings further implicate disruption of redox homeostasis as a key pathogenic mechanism in VLCAD deficiency and support the notion that a deranged thiol metabolism might be an important pathogenic factor in VLCAD deficiency.


Asunto(s)
Bezafibrato/farmacología , Síndromes Congénitos de Insuficiencia de la Médula Ósea/tratamiento farmacológico , Metabolismo Energético , Fibroblastos/efectos de los fármacos , Hipolipemiantes/farmacología , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Musculares/tratamiento farmacológico , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea/metabolismo , Síndromes Congénitos de Insuficiencia de la Médula Ósea/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Errores Innatos del Metabolismo Lipídico/metabolismo , Errores Innatos del Metabolismo Lipídico/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Estrés Oxidativo , Receptores Activados del Proliferador del Peroxisoma/genética
6.
Mol Genet Genomic Med ; 7(9): e915, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31392824

RESUMEN

BACKGROUND: Multiple acyl-CoA dehydrogenase deficiency (MADD), also known as glutaric aciduria type II, is a mitochondrial fatty acid oxidation disorder caused by variants in ETFA, ETFB, and ETFDH. Recently, riboflavin transporter genes and the mitochondrial FAD transporter gene have also been associated with MADD-like phenotype. METHODS: We present a case of MADD identified by newborn biochemical screening in a full-term infant suggestive of both medium-chain acyl-CoA dehydrogenase deficiency and MADD. Urine organic acid GC/MS analysis was also concerning for both disorders. However, panel sequencing of ETFA, ETFB, ETFDH, and ACADM was unrevealing. Ultimately, a variant in the FAD synthase gene, FLAD1 was found explaining the clinical presentation. RESULTS: Exome sequencing identified compound heterozygous variants in FLAD1: NM_025207.4: c.[442C>T];[1588C>T], p.[Arg148*];[Arg530Cys]. The protein damaging effects were confirmed by Western blot. The patient remained asymptomatic and there was no clinical decompensation during the first year of life. Plasma acylcarnitine and urinary organic acid analyses normalized without any treatment. Riboflavin supplementation was started at 15 months. CONCLUSION: Newborn screening, designed to screen for specific treatable congenital metabolic diseases, may also lead to the diagnosis of additional, very rare metabolic disorders such as FLAD1 deficiency. The case further illustrates that even milder forms of FLAD1 deficiency are detectable in the asymptomatic state by newborn screening.


Asunto(s)
Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Nucleotidiltransferasas/genética , Carnitina/análogos & derivados , Carnitina/sangre , Femenino , Humanos , Recién Nacido , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/sangre , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/orina , Mutación , Riboflavina/administración & dosificación
8.
Am J Hum Genet ; 98(6): 1130-1145, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259049

RESUMEN

Multiple acyl-CoA dehydrogenase deficiencies (MADDs) are a heterogeneous group of metabolic disorders with combined respiratory-chain deficiency and a neuromuscular phenotype. Despite recent advances in understanding the genetic basis of MADD, a number of cases remain unexplained. Here, we report clinically relevant variants in FLAD1, which encodes FAD synthase (FADS), as the cause of MADD and respiratory-chain dysfunction in nine individuals recruited from metabolic centers in six countries. In most individuals, we identified biallelic frameshift variants in the molybdopterin binding (MPTb) domain, located upstream of the FADS domain. Inasmuch as FADS is essential for cellular supply of FAD cofactors, the finding of biallelic frameshift variants was unexpected. Using RNA sequencing analysis combined with protein mass spectrometry, we discovered FLAD1 isoforms, which only encode the FADS domain. The existence of these isoforms might explain why affected individuals with biallelic FLAD1 frameshift variants still harbor substantial FADS activity. Another group of individuals with a milder phenotype responsive to riboflavin were shown to have single amino acid changes in the FADS domain. When produced in E. coli, these mutant FADS proteins resulted in impaired but detectable FADS activity; for one of the variant proteins, the addition of FAD significantly improved protein stability, arguing for a chaperone-like action similar to what has been reported in other riboflavin-responsive inborn errors of metabolism. In conclusion, our studies identify FLAD1 variants as a cause of potentially treatable inborn errors of metabolism manifesting with MADD and shed light on the mechanisms by which FADS ensures cellular FAD homeostasis.


Asunto(s)
Mutación del Sistema de Lectura/genética , Enfermedades Mitocondriales/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Nucleotidiltransferasas/genética , Riboflavina/farmacología , Complejo Vitamínico B/farmacología , Adulto , Western Blotting , Estudios de Casos y Controles , Células Cultivadas , Transporte de Electrón , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Flavina-Adenina Dinucleótido/metabolismo , Perfilación de la Expresión Génica , Humanos , Lactante , Recién Nacido , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Masculino , Enfermedades Mitocondriales/tratamiento farmacológico , Enfermedades Mitocondriales/patología , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/patología , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutagénesis Sitio-Dirigida , Unión Proteica , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Adulto Joven
9.
J Inherit Metab Dis ; 38(4): 703-19, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26025548

RESUMEN

Mitochondria play a key role in overall cell physiology and health by integrating cellular metabolism with cellular defense and repair mechanisms in response to physiological or environmental changes or stresses. In fact, dysregulation of mitochondrial stress responses and its consequences in the form of oxidative stress, has been linked to a wide variety of diseases including inborn errors of metabolism. In this review we will summarize how the functional state of mitochondria -- and especially the concentration of reactive oxygen species (ROS), produced in connection with the respiratory chain -- regulates cellular stress responses by redox regulation of nuclear gene networks involved in repair systems to maintain cellular homeostasis and health. Based on our own and other's studies we re-introduce the ROS triangle model and discuss how inborn errors of mitochondrial metabolism, by production of pathological amounts of ROS, may cause disturbed redox signalling and induce chronic cell stress with non-resolving or compromised cell repair responses and increased susceptibility to cell stress induced cell death. We suggest that this model may have important implications for those inborn errors of metabolism, where mitochondrial dysfunction plays a major role, as it allows the explanation of oxidative stress, metabolic reprogramming and altered signalling growth pathways that have been reported in many of the diseases. It is our hope that the model may facilitate novel ideas and directions that can be tested experimentally and used in the design of future new approaches for pre-symptomatic diagnosis and prognosis and perhaps more effective treatments of inborn errors of metabolism.


Asunto(s)
Errores Innatos del Metabolismo/metabolismo , Estrés Oxidativo/fisiología , Transducción de Señal/fisiología , Humanos , Errores Innatos del Metabolismo/fisiopatología , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Biogénesis de Organelos , Oxidación-Reducción
10.
JIMD Rep ; 23: 67-70, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25763512

RESUMEN

A neonate with low blood free carnitine level on newborn tandem mass spectrometry screening was evaluated for possible carnitine transporter defect (CTD). The plasma concentration of free carnitine was marginally reduced, and the concentrations of acylcarnitines (including C6, C8, and C10:1) were normal on confirmatory tests. Organic acids in urine were normal. In addition, none of the frequent Faroese SLC22A5 mutations (p.N32S, c.825-52G>A) which are common in the Danish population were identified. Evaluation of the mother showed low-normal free carnitine, but highly elevated medium-chain acylcarnitines (C6, C8, and C10:1) consistent with medium-chain acyl-CoA dehydrogenase deficiency (MCADD). The diagnosis was confirmed by the finding of homozygous presence of the c.985A>G mutation in ACADM.

11.
Hum Mol Genet ; 23(16): 4285-301, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24698980

RESUMEN

Mitochondrial dysfunction and oxidative stress are central to the molecular pathology of many human diseases. Riboflavin responsive multiple acyl-CoA dehydrogenation deficiency (RR-MADD) is in most cases caused by variations in the gene coding for electron transfer flavoprotein-ubiquinone oxidoreductase (ETF-QO). Currently, patients with RR-MADD are treated with high doses of riboflavin resulting in improvements of the clinical and biochemical profiles. However, in our recent studies of RR-MADD, we have shown that riboflavin treatment cannot fully correct the molecular defect in patient cells producing increased reactive oxygen species (ROS). In the current study, we aim to elucidate the cellular consequences of increased ROS by studying the cellular ROS adaption systems including antioxidant system, mitochondrial dynamics and metabolic reprogramming. We have included fibroblasts from six unrelated RR-MADD patients and two control fibroblasts cultivated under supplemented and depleted riboflavin conditions and with coenzyme Q10 (CoQ10) treatment. We demonstrated inhibition of mitochondrial fusion with increased fractionation and mitophagy in the patient fibroblasts. Furthermore, we indicated a shift in the energy metabolism by decreased protein levels of SIRT3 and decreased expression of fatty acid ß-oxidation enzymes in the patient fibroblasts. Finally, we showed that CoQ10 treatment has a positive effect on the mitochondrial dynamic in the patient fibroblasts, indicated by increased mitochondrial fusion marker and reduced mitophagy. In conclusion, our results indicate that RR-MADD patient fibroblasts suffer from a general mitochondria dysfunction, probably initiated as a rescue mechanism for the patient cells to escape apoptosis as a result of the oxidative stress.


Asunto(s)
Fibroblastos/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Estrés Oxidativo/efectos de los fármacos , Riboflavina/farmacología , Piel/metabolismo , Vitaminas/farmacología , Antioxidantes/metabolismo , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Humanos , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Piel/citología , Piel/efectos de los fármacos , Ubiquinona/análogos & derivados , Ubiquinona/farmacología
12.
Mol Genet Metab ; 111(3): 360-368, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24485985

RESUMEN

Short-chain acyl-CoA dehydrogenase (SCAD) deficiency is a rare inherited autosomal recessive disorder with not yet well established mechanisms of disease. In the present study, the mitochondrial proteome of five symptomatic patients homozygous for missense variations in the SCAD gene ACADS was investigated in an extensive large-scale proteomic study to map protein perturbations linked to the disease. Fibroblast cultures of patient cells homozygous for either c.319C>T/p.Arg107Cys (n=2) or c.1138C>T/p.Arg380Trp (n=3) in ACADS, and healthy controls (normal human dermal fibroblasts), were studied. The mitochondrial proteome derived from these cultures was analyzed by label free proteomics using high mass accuracy nanoliquid chromatography tandem mass spectrometry (nanoLC-MS/MS). More than 300 mitochondrial proteins were identified and quantified. Thirteen proteins had significant alteration in protein levels in patients carrying variation c.319C>T in ACADS compared to controls and they belonged to various pathways, such as the antioxidant system and amino acid metabolism. Twenty-two proteins were found significantly altered in patients carrying variation c.1138C>T which included proteins associated with fatty acid ß-oxidation, amino acid metabolism and protein quality control system. Three proteins were found significantly regulated in both patient groups: adenylate kinase 4 (AK4), nucleoside diphosphate kinase A (NME1) and aldehyde dehydrogenase family 4 member A1 (ALDH4A1). Proteins AK4 and NME1 deserve further investigation because of their involvement in energy reprogramming, cell survival and proliferation with relevance for SCAD deficiency and related metabolic disorders.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Butiril-CoA Deshidrogenasa/genética , Errores Innatos del Metabolismo Lipídico/genética , Mitocondrias/genética , Proteínas Mitocondriales/biosíntesis , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/metabolismo , Butiril-CoA Deshidrogenasa/metabolismo , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Errores Innatos del Metabolismo Lipídico/metabolismo , Errores Innatos del Metabolismo Lipídico/patología , Masculino , Mitocondrias/patología , Estrés Oxidativo/genética , Proteómica , Espectrometría de Masas en Tándem
13.
Hum Mutat ; 35(1): 86-95, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24123825

RESUMEN

Multiple acyl-CoA dehydrogenation deficiency is a disorder of fatty acid and amino acid oxidation caused by defects of electron transfer flavoprotein (ETF) or its dehydrogenase (ETFDH). A clear relationship between genotype and phenotype makes genotyping of patients important not only diagnostically but also for prognosis and for assessment of treatment. In the present study, we show that a predicted benign ETFDH missense variation (c.158A>G/p.Lys53Arg) in exon 2 causes exon skipping and degradation of ETFDH protein in patient samples. Using splicing reporter minigenes and RNA pull-down of nuclear proteins, we show that the c.158A>G variation increases the strength of a preexisting exonic splicing silencer (ESS) motif UAGGGA. This ESS motif binds splice inhibitory hnRNP A1, hnRNP A2/B1, and hnRNP H proteins. Binding of these inhibitory proteins prevents binding of the positive splicing regulatory SRSF1 and SRSF5 proteins to nearby and overlapping exonic splicing enhancer elements and this causes exon skipping. We further suggest that binding of hnRNP proteins to UAGGGA is increased by triggering synergistic hnRNP H binding to GGG triplets located upstream and downsteam of the UAGGGA motif. A number of disease-causing exonic elements that induce exon skipping in other genes have a similar architecture as the one in ETFDH exon 2.


Asunto(s)
Adenosina/metabolismo , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Guanina/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Empalme del ARN , Secuencias de Aminoácidos , Cadáver , Elementos de Facilitación Genéticos , Exones , Regulación de la Expresión Génica , Variación Genética , Células HEK293 , Células HeLa , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo F-H/metabolismo , Humanos , Recién Nacido , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/diagnóstico , Mutación Missense , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ADN , Factores de Empalme Serina-Arginina , Elementos Silenciadores Transcripcionales , Repeticiones de Trinucleótidos
14.
Hum Mol Genet ; 22(19): 3819-27, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23727839

RESUMEN

Coenzyme Q10 (CoQ10) is essential for the energy production of the cells and as an electron transporter in the mitochondrial respiratory chain. CoQ10 links the mitochondrial fatty acid ß-oxidation to the respiratory chain by accepting electrons from electron transfer flavoprotein-ubiquinone oxidoreductase (ETF-QO). Recently, it was shown that a group of patients with the riboflavin responsive form of multiple acyl-CoA dehydrogenation deficiency (RR-MADD) carrying inherited amino acid variations in ETF-QO also had secondary CoQ10 deficiency with beneficial effects of CoQ10 treatment, thus adding RR-MADD to an increasing number of diseases involving secondary CoQ10 deficiency. In this study, we show that moderately decreased CoQ10 levels in fibroblasts from six unrelated RR-MADD patients were associated with increased levels of mitochondrial reactive oxygen species (ROS). Treatment with CoQ10, but not with riboflavin, could normalize the CoQ10 level and decrease the level of ROS in the patient cells. Additionally, riboflavin-depleted control fibroblasts showed moderate CoQ10 deficiency, but not increased mitochondrial ROS, indicating that variant ETF-QO proteins and not CoQ10 deficiency are the causes of mitochondrial ROS production in the patient cells. Accordingly, the corresponding variant Rhodobacter sphaeroides ETF-QO proteins, when overexpressed in vitro, bind a CoQ10 pseudosubstrate, Q10Br, less tightly than the wild-type ETF-QO protein, suggesting that molecular oxygen can get access to the electrons in the misfolded ETF-QO protein, thereby generating superoxide and oxidative stress, which can be reversed by CoQ10 treatment.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Fibroblastos/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Estrés Oxidativo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Ubiquinona/análogos & derivados , Acilcoenzima A/metabolismo , Ataxia/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Cultivadas , Variación Genética , Humanos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/complicaciones , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Debilidad Muscular/metabolismo , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Rhodobacter sphaeroides/genética , Rhodobacter sphaeroides/metabolismo , Riboflavina/metabolismo , Riboflavina/farmacología , Ubiquinona/deficiencia , Ubiquinona/metabolismo , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
15.
Mol Genet Metab ; 107(3): 281-93, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22795865

RESUMEN

Expanded newborn screening for selected inborn errors of metabolism (IEM) in Denmark, the Faroe Islands and Greenland was introduced in 2002. We now present clinical, biochemical, and statistical results of expanded screening (excluding PKU) of 504,049 newborns during nine years as well as diagnoses and clinical findings in 82,930 unscreened newborns born in the same period. The frequencies of diagnoses made within the panel of disorders screened for are compared with the frequencies of the disorders in the decade preceding expanded newborn screening. The expanded screening was performed as a pilot study during the first seven years, and the experience obtained during these years was used in the development of the routine neonatal screening program introduced in 2009. Methods for screening included tandem mass spectrometry and an assay for determination of biotinidase activity. A total of 310 samples from 504,049 newborns gave positive screening results. Of the 310 results, 114 were true positive, including results from 12 newborns in which the disease in question was subsequently diagnosed in their mothers. Thus, the overall frequency of an IEM in the screening panel was 1:4942 (mothers excluded) or 1:4421 (mothers included). The false positive rate was 0.038% and positive predictive value 37%. Overall specificity was 99.99%. All patients with true positive results were followed in The Center for Inherited Metabolic Disorders in Copenhagen, and the mean follow-up period was 45 months (range 2109 months). There were no deaths among the 102 children, and 94% had no clinically significant sequelae at last follow-up. Our study confirms the higher frequency of selected IEM after implementation of expanded newborn screening and suggests an improved outcome for several disorders. We argue that newborn screening for these disorders should be standard of care, though unresolved issues remain, e.g. about newborns with a potential for remaining asymptomatic throughout life. Well organized logistics of the screening program from screening laboratory to centralized, clinical management is important.


Asunto(s)
Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/metabolismo , Tamizaje Neonatal/organización & administración , Biotinidasa/metabolismo , Niño , Dinamarca/epidemiología , Reacciones Falso Positivas , Femenino , Groenlandia/epidemiología , Humanos , Recién Nacido , Estudios Longitudinales , Masculino , Errores Innatos del Metabolismo/epidemiología , Proyectos Piloto , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem
16.
Hum Mol Genet ; 21(15): 3435-48, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22611163

RESUMEN

Riboflavin-responsive forms of multiple acyl-CoA dehydrogenation deficiency (RR-MADD) have been known for years, but with presumed defects in the formation of the flavin adenine dinucleotide (FAD) co-factor rather than genetic defects of electron transfer flavoprotein (ETF) or electron transfer flavoprotein-ubiquinone oxidoreductase (ETF-QO). It was only recently established that a number of RR-MADD patients carry genetic defects in ETF-QO and that the well-documented clinical efficacy of riboflavin treatment may be based on a chaperone effect that can compensate for inherited folding defects of ETF-QO. In the present study, we investigate the molecular mechanisms and the genotype-phenotype relationships for the riboflavin responsiveness in MADD, using a human HEK-293 cell expression system. We studied the influence of riboflavin and temperature on the steady-state level and the activity of variant ETF-QO proteins identified in patients with RR-MADD, or non- and partially responsive MADD. Our results showed that variant ETF-QO proteins associated with non- and partially responsive MADD caused severe misfolding of ETF-QO variant proteins when cultured in media with supplemented concentrations of riboflavin. In contrast, variant ETF-QO proteins associated with RR-MADD caused milder folding defects when cultured at the same conditions. Decreased thermal stability of the variants showed that FAD does not completely correct the structural defects induced by the variation. This may cause leakage of electrons and increased reactive oxygen species, as reflected by increased amounts of cellular peroxide production in HEK-293 cells expressing the variant ETF-QO proteins. Finally, we found indications of prolonged association of variant ETF-QO protein with the Hsp60 chaperonin in the mitochondrial matrix, supporting indications of folding defects in the variant ETF-QO proteins.


Asunto(s)
Flavoproteínas Transportadoras de Electrones/genética , Variación Genética , Proteínas Hierro-Azufre/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Riboflavina/metabolismo , Transporte de Electrón , Flavoproteínas Transportadoras de Electrones/metabolismo , Flavina-Adenina Dinucleótido/metabolismo , Células HEK293 , Humanos , Proteínas Hierro-Azufre/metabolismo , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/metabolismo , Mutación , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Pliegue de Proteína , Estructura Terciaria de Proteína , Transfección
17.
Hum Mutat ; 32(1): E1976-84, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21089064

RESUMEN

Riboflavin, or vitamin B2, is a precursor to flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN) molecules, required in biological oxidation-reduction reactions. We previously reported a case of a newborn female who had clinical and biochemical features of multiple acyl-CoA dehydrogenation deficiency (MADD), which was corrected by riboflavin supplementation. The mother was then found to be persistently riboflavin deficient, suggesting that a possible genetic defect in riboflavin transport in the mother was the cause of the transient MADD seen in the infant. Two recently-identified riboflavin transporters G protein-coupled receptor 172B (GPR172B or RFT1) and riboflavin transporter 2 (C20orf54 or RFT2) were screened for mutations. Two missense sequence variations, c.209A>G [p.Q70R] and c.886G>A [p.V296M] were found in GPR172B. In vitro functional studies of both missense variations showed that riboflavin transport was unaffected by these variations. Quantitative real-time PCR revealed a de novo deletion in GPR172B spanning exons 2 and 3 in one allele from the mother. We postulate that haploinsufficiency of this riboflavin transporter causes mild riboflavin deficiency, and when coupled with nutritional riboflavin deficiency in pregnancy, resulted in the transient riboflavin-responsive disease seen in her newborn infant. This is the first report of a genetic defect in riboflavin transport in humans.


Asunto(s)
Proteínas de Transporte de Membrana/genética , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/etiología , Receptores Acoplados a Proteínas G/genética , Deficiencia de Riboflavina/complicaciones , Deficiencia de Riboflavina/genética , Adulto , Variaciones en el Número de Copia de ADN , Exones , Femenino , Eliminación de Gen , Genotipo , Células HEK293 , Humanos , Recién Nacido , Masculino , Linaje
18.
J Inherit Metab Dis ; 34(1): 185-95, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21103935

RESUMEN

UNLABELLED: Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is a fatty acid oxidation disorder with especially high mortality and uncertain long-term outcome. The aim of the study was to analyze the influence of diagnostic approach on survival in 59 affected children. Referral to a metabolic center was replaced over time by urine/blood testing in centralized metabolic laboratory (selective screening) and by pilot tandem mass spectrometry newborn screening (NBS). Molecular analysis revealed the prevalent mutation in the HADHA gene in all 58 examined cases. Twenty patients died. The number of detections and number of deaths were respectively 9 and 4 (44%) in the patients recognized by differential diagnosis, 28 and 9 (32%) - by selective screening, and 11 and 1 (9%) - by NBS. In 80% of cases the death occurred before or within 3 weeks from the identification. Urgent and active metabolic service remarkably influenced the surviving. The current age of 39 survivors is 0.5 to 23 yrs (mean 7.2 yrs). The disease frequency estimated on the patients number was 1: 115 450, whereas in the pilot NBS - 1: 109 750 (658 492 neonates tested). Interestingly, the phenylalanine level in asymptomatic neonates frequently exceeded the cut-off values. CONCLUSIONS: 1) Urgent metabolic intervention decreases mortality of LCHAD-deficient patients, but the prognosis is still uncertain. 2) Emergent metabolic reporting and service are crucial also for the survival of neonates detected by NBS. 3) The nationwide selective screening appeared efficient in LCHADD detection in the country. 4) Transient mild hyperphenylalaninaemia may occur in LCHAD-deficient newborns.


Asunto(s)
Servicio de Urgencia en Hospital , Tamizaje Neonatal/métodos , Examen Físico/métodos , 3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , 3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , 3-Hidroxiacil-CoA Deshidrogenasas/orina , Adolescente , Carnitina/análogos & derivados , Carnitina/análisis , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN/métodos , Desecación , Servicio de Urgencia en Hospital/organización & administración , Femenino , Cromatografía de Gases y Espectrometría de Masas/métodos , Humanos , Lactante , Recién Nacido , Unidades de Cuidado Intensivo Neonatal/organización & administración , 3-Hidroxiacil-CoA Deshidrogenasa de Cadena Larga , Masculino , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/mortalidad , Errores Innatos del Metabolismo/orina , Tamizaje Neonatal/organización & administración , Proyectos Piloto , Polimorfismo de Longitud del Fragmento de Restricción , Sobrevida , Factores de Tiempo , Urinálisis/métodos
19.
J Inherit Metab Dis ; 33 Suppl 3: S373-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20814823

RESUMEN

Isolated long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) is associated with c.1528G>C substitution in the HADHA gene, since most patients have the prevalent mutation on at least one allele. As it is known that the disease is relatively frequent in Europe, especially around the Baltic Sea, and that the majority of Polish LCHADD patients originate from the coastal Pomeranian province, partly inhabited by an ancient ethnic group, the Kashubians, we aimed to determine the carrier frequency of the prevalent HADHA mutation in various districts of Poland with special focus on the Kashubian district. A total of 6,854 neonatal dried blood samples from the entire country, including 2,976 Pomeranian neonates of Kashubian origin, were c.1528G>C genotyped. Fifty-nine heterozygous carriers for the prevalent c.1528G>C substitution (41 Pomeranian children) were detected in the studied group. Our data reveal a geographically skewed distribution of the c.1528C allele in the Polish population; in the northern Pomeranian province the carrier frequency is 1:73, which is the highest frequency ever reported, whereas in the remaining regions it is 1:217. Hence, the incidence of LCHADD in Poland is predicted to be 1:118,336 versus 1:16,900 in the Pomeranian district. Despite the relative rarity of the disease, screening for LCHADD in neonates born in the northern part of Poland, especially those of Kashubian origin, is justified. Our data allow us to suggest a probable Kashubian origin of the prevalent c.1528G>C mutation.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , Cardiomiopatías/epidemiología , Cardiomiopatías/genética , Errores Innatos del Metabolismo Lipídico/epidemiología , Errores Innatos del Metabolismo Lipídico/genética , Miopatías Mitocondriales/epidemiología , Miopatías Mitocondriales/genética , Subunidad alfa de la Proteína Trifuncional Mitocondrial/deficiencia , Subunidad alfa de la Proteína Trifuncional Mitocondrial/genética , Mutación , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/genética , Rabdomiólisis/epidemiología , Rabdomiólisis/genética , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Cardiomiopatías/diagnóstico , Cardiomiopatías/enzimología , Análisis Mutacional de ADN , Pruebas con Sangre Seca , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Heterocigoto , Humanos , Recién Nacido , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/enzimología , Miopatías Mitocondriales/diagnóstico , Miopatías Mitocondriales/enzimología , Proteína Trifuncional Mitocondrial/deficiencia , Tamizaje Neonatal/métodos , Enfermedades del Sistema Nervioso/diagnóstico , Enfermedades del Sistema Nervioso/enzimología , Fenotipo , Polonia/epidemiología , Valor Predictivo de las Pruebas , Prevalencia , Características de la Residencia , Rabdomiólisis/diagnóstico , Rabdomiólisis/enzimología
20.
J Inherit Metab Dis ; 33(5): 547-53, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20151199

RESUMEN

In fatty acid oxidation defects, the majority of gene variations are of the missense type and, therefore, prone to inducing misfolding in the resulting mutant protein. The fate of the mutant protein depends on the nature of the gene variation and other genetic factors as well as cellular and environmental factors. Since it has been shown that certain fatty acid oxidation enzyme proteins, exemplified by mutant medium-chain and short-chain acyl-CoA dehydrogenases as well as electron transfer flavoprotein and electron transfer flavoprotein dehydrogenase, may accumulate during cellular stress, e.g. elevated temperature, there is speculation about how such proteins may disturb the integrity of the putative fatty acid oxidation metabolone, in which the two flavoproteins link the matrix-located acyl-CoA dehydrogenases to the respiratory chain in the mitochondrial inner membrane. However, since studies so far have not been able to define the fatty acid oxidation metabolone, it is concluded that new concepts and refined techniques are required to answer these questions and thereby contribute to the elucidation of the cellular pathophysiology and the genotype-phenotype relationship in fatty acid oxidation defects.


Asunto(s)
Metabolismo Energético , Ácidos Grasos/metabolismo , Errores Innatos del Metabolismo Lipídico/enzimología , Mitocondrias/enzimología , Enfermedades Mitocondriales/enzimología , Animales , Metabolismo Energético/genética , Genotipo , Humanos , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/fisiopatología , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/fisiopatología , Oxidación-Reducción , Fenotipo , Conformación Proteica , Pliegue de Proteína , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA