Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(17): 2717-2734, 2023 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-37369025

RESUMEN

Inherited disorders of mitochondrial metabolism, including isolated methylmalonic aciduria, present unique challenges to energetic homeostasis by disrupting energy-producing pathways. To better understand global responses to energy shortage, we investigated a hemizygous mouse model of methylmalonyl-CoA mutase (Mmut)-type methylmalonic aciduria. We found Mmut mutant mice to have reduced appetite, energy expenditure and body mass compared with littermate controls, along with a relative reduction in lean mass but increase in fat mass. Brown adipose tissue showed a process of whitening, in line with lower body surface temperature and lesser ability to cope with cold challenge. Mutant mice had dysregulated plasma glucose, delayed glucose clearance and a lesser ability to regulate energy sources when switching from the fed to fasted state, while liver investigations indicated metabolite accumulation and altered expression of peroxisome proliferator-activated receptor and Fgf21-controlled pathways. Together, these shed light on the mechanisms and adaptations behind energy imbalance in methylmalonic aciduria and provide insight into metabolic responses to chronic energy shortage, which may have important implications for disease understanding and patient management.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Ratones , Animales , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Metabolismo Energético/genética , Hígado/metabolismo
2.
Circulation ; 137(24): 2592-2608, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29353241

RESUMEN

BACKGROUND: Nutrients are transported through endothelial cells before being metabolized in muscle cells. However, little is known about the regulation of endothelial transport processes. Notch signaling is a critical regulator of metabolism and angiogenesis during development. Here, we studied how genetic and pharmacological manipulation of endothelial Notch signaling in adult mice affects endothelial fatty acid transport, cardiac angiogenesis, and heart function. METHODS: Endothelial-specific Notch inhibition was achieved by conditional genetic inactivation of Rbp-jκ in adult mice to analyze fatty acid metabolism and heart function. Wild-type mice were treated with neutralizing antibodies against the Notch ligand Delta-like 4. Fatty acid transport was studied in cultured endothelial cells and transgenic mice. RESULTS: Treatment of wild-type mice with Delta-like 4 neutralizing antibodies for 8 weeks impaired fractional shortening and ejection fraction in the majority of mice. Inhibition of Notch signaling specifically in the endothelium of adult mice by genetic ablation of Rbp-jκ caused heart hypertrophy and failure. Impaired heart function was preceded by alterations in fatty acid metabolism and an increase in cardiac blood vessel density. Endothelial Notch signaling controlled the expression of endothelial lipase, Angptl4, CD36, and Fabp4, which are all needed for fatty acid transport across the vessel wall. In endothelial-specific Rbp-jκ-mutant mice, lipase activity and transendothelial transport of long-chain fatty acids to muscle cells were impaired. In turn, lipids accumulated in the plasma and liver. The attenuated supply of cardiomyocytes with long-chain fatty acids was accompanied by higher glucose uptake, increased concentration of glycolysis intermediates, and mTOR-S6K signaling. Treatment with the mTOR inhibitor rapamycin or displacing glucose as cardiac substrate by feeding a ketogenic diet prolonged the survival of endothelial-specific Rbp-jκ-deficient mice. CONCLUSIONS: This study identifies Notch signaling as a novel regulator of fatty acid transport across the endothelium and as an essential repressor of angiogenesis in the adult heart. The data imply that the endothelium controls cardiomyocyte metabolism and function.


Asunto(s)
Endotelio Vascular/metabolismo , Ácidos Grasos/metabolismo , Miocardio/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Remodelación Vascular , Proteínas Adaptadoras Transductoras de Señales , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Animales , Antígenos CD36/genética , Antígenos CD36/metabolismo , Proteínas de Unión al Calcio , Endotelio Vascular/citología , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Ácidos Grasos/genética , Glucosa/genética , Glucosa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Neovascularización Fisiológica , Receptores Notch/genética , Proteínas Quinasas S6 Ribosómicas/genética , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
3.
Am J Hum Genet ; 99(2): 414-22, 2016 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-27426735

RESUMEN

tRNA synthetase deficiencies are a growing group of genetic diseases associated with tissue-specific, mostly neurological, phenotypes. In cattle, cytosolic isoleucyl-tRNA synthetase (IARS) missense mutations cause hereditary weak calf syndrome. Exome sequencing in three unrelated individuals with severe prenatal-onset growth retardation, intellectual disability, and muscular hypotonia revealed biallelic mutations in IARS. Studies in yeast confirmed the pathogenicity of identified mutations. Two of the individuals had infantile hepatopathy with fibrosis and steatosis, leading in one to liver failure in the course of infections. Zinc deficiency was present in all affected individuals and supplementation with zinc showed a beneficial effect on growth in one.


Asunto(s)
Alelos , Retardo del Crecimiento Fetal/genética , Discapacidad Intelectual/genética , Isoleucina-ARNt Ligasa/genética , Hepatopatías/congénito , Hepatopatías/genética , Hipotonía Muscular/congénito , Hipotonía Muscular/genética , Mutación , Adolescente , Animales , Niño , Preescolar , Suplementos Dietéticos , Hígado Graso/genética , Femenino , Fibrosis/genética , Humanos , Lactante , Recién Nacido , Isoleucina-ARNt Ligasa/deficiencia , Fallo Hepático/genética , Masculino , Síndrome , Pez Cebra/genética , Zinc/administración & dosificación , Zinc/deficiencia , Zinc/uso terapéutico
4.
J Inherit Metab Dis ; 41(1): 91-99, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29098534

RESUMEN

Glutaric aciduria type I is a rare, autosomal recessive, inherited defect of glutaryl-CoA dehydrogenase. Deficiency of this protein in L-lysine degradation leads to the characteristic accumulation of nontoxic glutarylcarnitine and neurotoxic glutaric acid (GA), glutaryl-CoA, and 3-hydroxyglutaric acid. Untreated patients develop bilateral lesions of basal ganglia resulting in a complex movement disorder with predominant dystonia in infancy and early childhood. The current pathomechanistic concept strongly focuses on imbalanced neuronal energy metabolism due to accumulating metabolites, whereas little is known about the pathomechanistic role of astrocytes, which are thought to be in constant metabolic crosstalk with neurons. We found that glutaric acid (GA) causes astrocytic cell death under starvation cell culture conditions, i.e. low glucose, without glutamine and fetal calf serum. Glutamine completely abolished GA-induced toxicity, suggesting involvement of glutaminolysis. Increasing dependence on glutaminolysis by chemical induction of hypoxia signaling-potentiated GA-induced toxicity. We further show that GA disturbs glutamine degradation by specifically inhibiting glutamate dehydrogenase. Summarizing our study shows that pathologically relevant concentrations of GA block an important step in the metabolic crosstalk between neurons and astrocytes, ultimately leading to astrocytic cell death.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Astrocitos/efectos de los fármacos , Encefalopatías Metabólicas/metabolismo , Inhibidores Enzimáticos/toxicidad , Glutamato Deshidrogenasa/antagonistas & inhibidores , Glutamina/metabolismo , Glutaratos/toxicidad , Glutaril-CoA Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/patología , Animales , Astrocitos/metabolismo , Astrocitos/patología , Encefalopatías Metabólicas/patología , Muerte Celular/efectos de los fármacos , Hipoxia de la Célula , Línea Celular , Glutamato Deshidrogenasa/metabolismo , Glutamina/farmacología , Glutaril-CoA Deshidrogenasa/metabolismo , Ratas
5.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2220-2228, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28545977

RESUMEN

Glutaric aciduria type I (GA-I) is a rare organic aciduria caused by the autosomal recessive inherited deficiency of glutaryl-CoA dehydrogenase (GCDH). GCDH deficiency leads to disruption of l-lysine degradation with characteristic accumulation of glutarylcarnitine and neurotoxic glutaric acid (GA), glutaryl-CoA, 3-hydroxyglutaric acid (3-OHGA). DHTKD1 acts upstream of GCDH, and its deficiency leads to none or often mild clinical phenotype in humans, 2-aminoadipic 2-oxoadipic aciduria. We hypothesized that inhibition of DHTKD1 may prevent the accumulation of neurotoxic dicarboxylic metabolites suggesting DHTKD1 inhibition as a possible treatment strategy for GA-I. In order to validate this hypothesis we took advantage of an existing GA-I (Gcdh-/-) mouse model and established a Dhtkd1 deficient mouse model. Both models reproduced the biochemical and clinical phenotype observed in patients. Under challenging conditions of a high lysine diet, only Gcdh-/- mice but not Dhtkd1-/- mice developed clinical symptoms such as lethargic behaviour and weight loss. However, the genetic Dhtkd1 inhibition in Dhtkd1-/-/Gcdh-/- mice could not rescue the GA-I phenotype. Biochemical results confirm this finding with double knockout mice showing similar metabolite accumulations as Gcdh-/- mice with high GA in brain and liver. This suggests that DHTKD1 inhibition alone is not sufficient to treat GA-I, but instead a more complex strategy is needed. Our data highlights the many unresolved questions within the l-lysine degradation pathway and provides evidence for a so far unknown mechanism leading to glutaryl-CoA.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas/metabolismo , Encéfalo/metabolismo , Glutaratos/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Cetona Oxidorreductasas/deficiencia , Hígado/metabolismo , Lisina/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Animales , Encéfalo/patología , Encefalopatías Metabólicas/genética , Encefalopatías Metabólicas/terapia , Modelos Animales de Enfermedad , Glutaril-CoA Deshidrogenasa/genética , Glutaril-CoA Deshidrogenasa/metabolismo , Cetona Oxidorreductasas/metabolismo , Hígado/patología , Ratones , Ratones Noqueados
6.
RNA ; 21(11): 1873-84, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26370583

RESUMEN

Mitochondrial RNA processing is an essential step for the synthesis of the components of the electron transport chain in all eukaryotic organisms, yet several aspects of mitochondrial RNA biogenesis and regulation are not sufficiently understood. RNA interactome capture identified several disease-relevant RNA-binding proteins (RBPs) with noncanonical RNA-binding architectures, including all six members of the FASTK (FAS-activated serine/threonine kinase) family of proteins. A mutation within one of these newly assigned FASTK RBPs, FASTKD2, causes a rare form of Mendelian mitochondrial encephalomyopathy. To investigate whether RNA binding of FASTKD2 contributes to the disease phenotype, we identified the RNA targets of FASTKD2 by iCLIP. FASTKD2 interacts with a defined set of mitochondrial transcripts including 16S ribosomal RNA (RNR2) and NADH dehydrogenase subunit 6 (ND6) messenger RNA. CRISPR-mediated deletion of FASTKD2 leads to aberrant processing and expression of RNR2 and ND6 mRNA that encodes a subunit of the respiratory complex I. Metabolic phenotyping of FASTKD2-deficient cells reveals impaired cellular respiration with reduced activities of all respiratory complexes. This work identifies key aspects of the molecular network of a previously uncharacterized, disease-relevant RNA-binding protein, FASTKD2, by a combination of genomic, molecular, and metabolic analyses.


Asunto(s)
Proteínas Mitocondriales/genética , Biosíntesis de Proteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al ARN/genética , ARN/genética , Línea Celular , Respiración de la Célula/genética , Células HEK293 , Humanos , Mitocondrias/genética , NADH Deshidrogenasa/genética , ARN Mensajero/genética , ARN Mitocondrial , ARN Ribosómico 16S/genética
7.
Biochim Biophys Acta ; 1852(5): 768-77, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25558815

RESUMEN

Glutaric aciduria type I is an inherited defect in L-lysine, L-hydroxylysine and L-tryptophan degradation caused by deficiency of glutaryl-CoA dehydrogenase (GCDH). The majority of untreated patients presents with accumulation of neurotoxic metabolites - glutaric acid (GA) and 3-hydroxyglutaric acid (3-OHGA) - and striatal injury. Gcdh(-/-) mice display elevated levels of GA and 3-OH-GA but do not spontaneously develop striatal lesions. L-lysine-enriched diets (appr. 235 mg/d) were suggested to induce a neurological phenotype similar to affected patients. In our hands 93% of mice stressed according to the published protocol remained asymptomatic. To understand the underlying mechanism, we modified their genetic background (F1 C57BL6/Jx129/SvCrl) and increased the daily oral L-lysine supply (235-433 mg). We identified three modulating factors, (1) gender, (2) genetic background, and (3) amount of L-lysine. Male mice displayed higher vulnerability and inbreeding for more than two generations as well as elevating L-lysine supply increased the diet-induced mortality rate (up to 89%). Onset of first symptoms leads to strongly reduced intake of food and, thus, L-lysine suggesting a threshold for toxic metabolite production to induce neurological disease. GA and 3-OH-GA tissue concentrations did not correlate with dietary L-lysine supply but differed between symptomatic and asymptomatic mice. Cerebral activities of glyceraldehyde 3-phosphate dehydrogenase, 2-oxoglutarate dehydrogenase complex, and aconitase were decreased. Symptomatic mice did not develop striatal lesions or intracerebral hemorrhages. We found severe spongiosis in the hippocampus of Gcdh(-/-) mice which was independent of dietary L-lysine supply. In conclusion, the L-lysine-induced pathology in Gcdh(-/-) mice depends on genetic and dietary parameters.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Encefalopatías Metabólicas/genética , Predisposición Genética a la Enfermedad/genética , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Lisina/administración & dosificación , Aconitato Hidratasa/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/etiología , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Encefalopatías Metabólicas/etiología , Encefalopatías Metabólicas/metabolismo , Dieta , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Metabolismo Energético/efectos de los fármacos , Femenino , Predisposición Genética a la Enfermedad/etiología , Glutaratos/metabolismo , Glutaril-CoA Deshidrogenasa/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Complejo Cetoglutarato Deshidrogenasa/metabolismo , Lisina/efectos adversos , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Riesgo , Factores Sexuales , Espectrofotometría
8.
J Inherit Metab Dis ; 39(5): 625-632, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27139199

RESUMEN

BACKGROUND: Hydroxyprolinemia is an inborn error of amino acid degradation that is considered a non-disease. Known for more than 50 years, its genetic cause and prevalence have remained unclear. In MS/MS newborn screening, the mass spectrum of hydroxyproline cannot be differentiated from isoleucine and leucine causing false positive newborn screening test results for maple syrup urine disease (MSUD). METHODS: We studied two siblings with hydroxyprolinemia via exome sequencing and confirmed the candidate gene in five further individuals with hydroxyprolinemia, who were all characterized biochemically and clinically. The prevalence was calculated based on the number of individuals with hydroxyprolinemia detected via MS/MS newborn screening at our centre from 2003 to 2014. RESULTS: In six cases, we identified homozygous or compound heterozygous mutations in PRODH2 as the underlying genetic cause of hydroxyprolinemia. One individual was heterozygous for a deletion in PRODH2 and had an intermittent biochemical phenotype with partial normalization of hydroxyproline concentrations. In one further individual with persistent hydroxyprolinemia no mutation in PRODH2 was found, raising the possibility of another defect of hydroxyproline degradation yet to be identified as the underlying cause of hydroxyprolinemia. Plasma hydroxyproline concentrations were clearly elevated in all individuals with biallelic mutations in PRODH2. All studied individuals remained asymptomatic, giving further evidence that hydroxyprolinemia is a benign condition. The estimated prevalence of hydroxyprolinemia in Germany is about one in 47,300 newborns. CONCLUSION: Our results establish mutations in PRODH2 as a cause of human hydroxyprolinemia via impaired dehydrogenation of hydroxyproline to delta1-pyroline-3-hydroxy-5-carboxylic acid, and we suggest PRODH2 be renamed HYPDH. Hydroxyprolinemia is an autosomal-recessively inherited benign condition. It is a frequent cause of false positive screening results for MSUD, the prevalence being about 2.5 times higher than that of MSUD.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/etiología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Mutación/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/deficiencia , Niño , Preescolar , Femenino , Alemania , Heterocigoto , Homocigoto , Humanos , Hidroxiprolina/genética , Lactante , Recién Nacido , Masculino , Enfermedad de la Orina de Jarabe de Arce/etiología , Enfermedad de la Orina de Jarabe de Arce/genética , Tamizaje Neonatal/métodos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Fenotipo , Prevalencia , Prolina Oxidasa/genética
9.
Am J Hum Genet ; 91(6): 1082-7, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23141293

RESUMEN

Abnormalities in metabolite profiles are valuable indicators of underlying pathologic conditions at the molecular level. However, their interpretation relies on detailed knowledge of the pathways, enzymes, and genes involved. Identification and characterization of their physiological function are therefore crucial for our understanding of human disease: they can provide guidance for therapeutic intervention and help us to identify suitable biomarkers for monitoring associated disorders. We studied two individuals with 2-aminoadipic and 2-oxoadipic aciduria, a metabolic condition that is still unresolved at the molecular level. This disorder has been associated with varying neurological symptoms. Exome sequencing of a single affected individual revealed compound heterozygosity for an initiating methionine mutation (c.1A>G) and a missense mutation (c.2185G>A [p.Gly729Arg]) in DHTKD1. This gene codes for dehydrogenase E1 and transketolase domain-containing protein 1, which is part of a 2-oxoglutarate-dehydrogenase-complex-like protein. Sequence analysis of a second individual identified the same missense mutation together with a nonsense mutation (c.1228C>T [p.Arg410(∗)]) in DHTKD1. Increased levels of 2-oxoadipate in individual-derived fibroblasts normalized upon lentiviral expression of the wild-type DHTKD1 mRNA. Moreover, investigation of L-lysine metabolism showed an accumulation of deuterium-labeled 2-oxoadipate only in noncomplemented cells, demonstrating that DHTKD1 codes for the enzyme mediating the last unresolved step in the L-lysine-degradation pathway. All together, our results establish mutations in DHTKD1 as a cause of human 2-aminoadipic and 2-oxoadipic aciduria via impaired turnover of decarboxylation 2-oxoadipate to glutaryl-CoA.


Asunto(s)
Ácido 2-Aminoadípico/orina , Adipatos/orina , Errores Innatos del Metabolismo de los Aminoácidos/genética , Cetona Oxidorreductasas/genética , Mutación , Alelos , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Secuencia de Aminoácidos , Exones , Femenino , Fibroblastos/metabolismo , Orden Génico , Genotipo , Humanos , Complejo Cetoglutarato Deshidrogenasa , Linaje , Fenotipo
10.
J Inherit Metab Dis ; 38(2): 265-72, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25214427

RESUMEN

Inherited deficiencies of the L-lysine catabolic pathway cause glutaric aciduria type I and pyridoxine-dependent epilepsy. Dietary modulation of cerebral L-lysine metabolism is thought to be an important therapeutic intervention for these diseases. To better understand cerebral L-lysine degradation, we studied in mice the two known catabolic routes -- pipecolate and saccharopine pathways -- using labeled stable L-lysine and brain peroxisomes purified according to a newly established protocol. Experiments with labeled stable L-lysine show that cerebral L-pipecolate is generated along two pathways: i) a minor proportion retrograde after ε-deamination of L-lysine along the saccharopine pathway, and ii) a major proportion anterograde after α-deamination of L-lysine along the pipecolate pathway. In line with these findings, we observed only little production of saccharopine in the murine brain. L-pipecolate oxidation was only detectable in brain peroxisomes, but L-pipecolate oxidase activity was low (7 ± 2µU/mg protein). In conclusion, L-pipecolate is a major degradation product from L-lysine in murine brain generated by α-deamination of this amino acid.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Encefalopatías Metabólicas/enzimología , Encefalopatías Metabólicas/genética , Encéfalo/enzimología , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/genética , Lisina/metabolismo , Ácidos Pipecólicos/metabolismo , Animales , Desaminación , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Hígado/enzimología , Lisina/análogos & derivados , Ratones Noqueados , Oxidación-Reducción , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Peroxisomas/enzimología , Fenotipo
11.
Biochim Biophys Acta ; 1823(5): 1041-52, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22429591

RESUMEN

Mitochondrial reactive oxygen species (ROS) are indispensible for T cell activation-induced expression of interleukin 2 (IL-2) and CD95 ligand (CD95L, FasL/Apo-1L) genes, and in turn, for CD95L-mediated activation-induced cell death (AICD). Here, we show that manganese superoxide dismutase (MnSOD/SOD2), a major mitochondrial antioxidative enzyme, constitutes an important control switch in the process of activation-induced oxidative signal generation in T cells. Analysis of the kinetics of T cell receptor (TCR)-triggered ROS production revealed a temporal association between higher MnSOD abundance/activity and a shut-down phase of oxidative signal generation. Transient or inducible MnSOD overexpression abrogated T cell activation-triggered mitochondrial ROS production as well as NF-κB- and AP-1-mediated transcription. Consequently, lowered expression of IL-2 and CD95L genes resulted in decreased IL-2 secretion and CD95L-dependent AICD. Moreover, upregulation of the mitochondrial MnSOD level is dependent on oxidation-sensitive transcription and not on the increase of mitochondrial mass. Thus, MnSOD-mediated negative feedback regulation of activation-induced mitochondrial ROS generation exemplifies a process of retrograde mitochondria-to-nucleus communication. Our finding underlines the critical role for MnSOD and mitochondria in the regulation of human T cell activation.


Asunto(s)
Activación de Linfocitos/inmunología , Transducción de Señal/inmunología , Superóxido Dismutasa/metabolismo , Linfocitos T/citología , Linfocitos T/enzimología , Muerte Celular/inmunología , Proteína Ligando Fas/metabolismo , Regulación de la Expresión Génica , Humanos , Células Jurkat , Mitocondrias/metabolismo , Modelos Inmunológicos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/genética , Linfocitos T/inmunología , Transcripción Genética
12.
J Inherit Metab Dis ; 36(4): 635-44, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23512157

RESUMEN

This review focuses on the pathophysiology of organic acidurias (OADs), in particular, OADs caused by deficient amino acid metabolism. OADs are termed classical if patients present with acute metabolic decompensation and multiorgan dysfunction or cerebral if patients predominantly present with neurological symptoms but without metabolic crises. In both groups, however, the brain is the major target. The high energy demand of the brain, the gate-keeping function of the blood-brain barrier, a high lipid content, vulnerable neuronal subpopulations, and glutamatergic neurotransmission all make the brain particularly vulnerable against mitochondrial dysfunction, oxidative stress, and excitotoxicity. In fact, toxic metabolites in OADs are thought to cause secondary impairment of energy metabolism; some of these toxic metabolites are trapped in the brain. In contrast to cerebral OADs, patients with classical OADs have an increased risk of multiorgan dysfunction. The lack of the anaplerotic propionate pathway, synergistic inhibition of energy metabolism by toxic metabolites, and multiple oxidative phosphorylation (OXPHOS) deficiency may best explain the involvement of organs with a high energy demand. Intriguingly, late-onset organ dysfunction may manifest even under metabolically stable conditions. This might be explained by chronic mitochondrial DNA depletion, increased production of reactive oxygen species, and altered gene expression due to histone modification. In conclusion, pathomechanisms underlying the acute disease manifestation in OADs, with a particular focus on the brain, are partially understood. More work is required to predict the risk and to elucidate the mechanism of late-onset organ dysfunction, extracerebral disease manifestation, and tumorigenesis.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas Innatas/metabolismo , Encéfalo/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/patología , Animales , Encéfalo/patología , Encefalopatías Metabólicas Innatas/patología , Metabolismo Energético , Humanos
13.
Pediatr Nephrol ; 28(2): 227-35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22814947

RESUMEN

Methylmalonic acidurias are a heterogeneous group of inborn errors of branched-chain amino acid metabolism. Depending on the underlying etiology, acute or chronic renal disease constitutes major (long-term) complications. In recent decades, overall survival has improved due to optimized treatment strategies based on the use of standardized emergency protocols and dialysis techniques. The majority of these patients, especially those having mut°, cblB, and cblA deficiency, are at increased risk of developing chronic kidney disease secondary to tubulointerstitial nephritis to require hemo- or peritoneal dialysis. Kidney and/or liver transplantation, as organ replacement, or even gene therapy on a limited scale, are controversially discussed treatment options in methylmalonic acidurias. The pathophysiological basis of renal disease has not been clarified in detail until now, but a severe mitochondrial dysfunction and an impairment of tubular dicarboxylic acid transport due to accumulated toxic metabolic compounds has been recently proposed. Another severe renal complication of methylmalonic acidurias is the occurrence of cblC-associated infantile atypical hemolytic syndrome, which can result in acute kidney injury. Close collaboration between (pediatric) nephrologists and metabolic specialists is required for the long-term management of these patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/complicaciones , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Ácido Metilmalónico/metabolismo , Insuficiencia Renal Crónica/etiología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Dieta , Suplementos Dietéticos , Genotipo , Humanos , Trasplante de Riñón , Mitocondrias/metabolismo , Fenotipo , Terapia de Reemplazo Renal
14.
Biochim Biophys Acta ; 1812(12): 1607-15, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21920437

RESUMEN

Wilson disease (WD) is caused by mutations of the WD gene ATP7B resulting in copper accumulation in different tissues. WD patients display hepatic and neurological disease with yet poorly understood pathomechanisms. Therefore, we studied age-dependent (3, 6, 47weeks) biochemical and bioenergetical changes in Atp7b(-/-) mice focusing on liver and brain. Mutant mice showed strongly elevated copper and iron levels. Age-dependently decreasing hepatic reduced glutathione levels along with increasing oxidized to reduced glutathione ratios in liver and brain of 47weeks old mice as well as elevated hepatic and cerebral superoxide dismutase activities in 3weeks old mutant mice highlighted oxidative stress in the investigated tissues. We could not find evidence that amino acid metabolism or beta-oxidation is impaired by deficiency of ATP7B. In contrast, sterol metabolism was severely dysregulated. In brains of 3week old mice cholesterol, 8-dehydrocholesterol, desmosterol, 7-dehydrocholesterol, and lathosterol were all highly increased. These changes reversed age-dependently resulting in reduced levels of all previously increased sterol metabolites in 47weeks old mice. A similar pattern of sterol metabolite changes was found in hepatic tissue, though less pronounced. Moreover, mitochondrial energy production was severely affected. Respiratory chain complex I activity was increased in liver and brain of mutant mice, whereas complex II, III, and IV activities were reduced. In addition, aconitase activity was diminished in brains of Atp7b(-/-) mice. Summarizing, our study reveals oxidative stress along with severe dysfunction of mitochondrial energy production and of sterol metabolism in Atp7b(-/-) mice shedding new light on the pathogenesis of WD.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Colesterol/metabolismo , Degeneración Hepatolenticular/metabolismo , Aconitato Hidratasa/metabolismo , Alanina/metabolismo , Animales , Encéfalo/enzimología , Encéfalo/metabolismo , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/metabolismo , Colesterol/sangre , Cobre/metabolismo , ATPasas Transportadoras de Cobre , Modelos Animales de Enfermedad , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Técnicas de Inactivación de Genes , Glutatión/metabolismo , Degeneración Hepatolenticular/sangre , Humanos , Hígado/enzimología , Hígado/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Mitocondrias/metabolismo , Estrés Oxidativo , Superóxido Dismutasa/metabolismo
15.
Langmuir ; 28(22): 8399-407, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22616616

RESUMEN

New triphenylene-based silanes 2-(ω-(chlorodimethylsilyl)-n-alkyl)-3,6,7,10,11-penta-m-alkoxytriphenylene 4 (Tm-Cn) with n = 8 or 9 and m = 7, 8, 9, 10, or 11 were synthesized, and their self-assembly behavior in the liquid state and at glass and silicon oxide surfaces was investigated. The mesomorphic properties of triphenylene silanes 4 (Tm-Cn) and their precursors 3 (Tm-Cn) were determined by differential scanning calorimetry (DSC), polarizing optical microscopy (POM), and X-ray diffraction. From the small-angle X-ray scattering (SAXS) regime, a preferential discotic lamellar mesophase can be deduced, and wide-angle X-ray scattering (WAXS) highlights the liquid-like characteristics of the alkyl side chains. To transfer these bulk structural properties to thin films, self-assembled monolayers (SAMs) were obtained by adsorption from solution and characterized by water contact angle measurements, null ellipsometry, and atomic force microscopy (AFM). Employing the concentration as an additional degree of freedom, binary SAMs of 2-(ω-(chlorodimethylsilyl)-undecyl)-3,6,7,10,11-penta-decyloxytriphenylene 4 (T10-C11) were coassembled with chlorodecyldimethylsilane or chlorodimethyloctadecylsilane, and their capability as model systems for organic templating was evaluated. The structure of the resulting binary mixed SAMs was analyzed by water contact angle measurements, null ellipsometry, and X-ray reflectivity (XRR) in combination with theoretical modeling by a multidimensional Parratt algorithm and AFM. The composition dependence of film thickness and roughness can be explained by a microscopic model including the steric hindrance of the respective molecular constituents.

16.
J Immunol ; 184(9): 4827-41, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20335530

RESUMEN

This article shows that T cell activation-induced expression of the cytokines IL-2 and -4 is determined by an oxidative signal originating from mitochondrial respiratory complex I. We also report that ciprofloxacin, a fluoroquinolone antibiotic, exerts immunosuppressive effects on human T cells suppressing this novel mechanism. Sustained treatment of preactivated primary human T cells with ciprofloxacin results in a dose-dependent inhibition of TCR-induced generation of reactive oxygen species (ROS) and IL-2 and -4 expression. This is accompanied by the loss of mitochondrial DNA and a resulting decrease in activity of the complex I. Consequently, using a complex I inhibitor or small interfering RNA-mediated downregulation of the complex I chaperone NDUFAF1, we demonstrate that TCR-triggered ROS generation by complex I is indispensable for activation-induced IL-2 and -4 expression and secretion in resting and preactivated human T cells. This oxidative signal (H(2)O(2)) synergizes with Ca(2+) influx for IL-2/IL-4 expression and facilitates induction of the transcription factors NF-kappaB and AP-1. Moreover, using T cells isolated from patients with atopic dermatitis, we show that inhibition of complex I-mediated ROS generation blocks disease-associated spontaneous hyperexpression and TCR-induced expression of IL-4. Prolonged ciprofloxacin treatment of T cells from patients with atopic dermatitis also blocks activation-induced expression and secretion of IL-4. Thus, our work shows that the activation phenotype of T cells is controlled by a mitochondrial complex I-originated oxidative signal.


Asunto(s)
Ciprofloxacina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Inmunosupresores/farmacología , Interleucina-2/genética , Interleucina-4/genética , Activación de Linfocitos/inmunología , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/inmunología , Células Cultivadas , ADN Mitocondrial/antagonistas & inhibidores , ADN Mitocondrial/genética , Dermatitis Atópica/enzimología , Dermatitis Atópica/genética , Dermatitis Atópica/inmunología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/genética , Regulación de la Expresión Génica/inmunología , Humanos , Inmunofenotipificación , Interleucina-2/antagonistas & inhibidores , Interleucina-2/metabolismo , Interleucina-4/antagonistas & inhibidores , Interleucina-4/metabolismo , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , NADH Deshidrogenasa/antagonistas & inhibidores , NADH Deshidrogenasa/genética , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/biosíntesis , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología , Células Th2/efectos de los fármacos , Células Th2/enzimología , Células Th2/inmunología , Factores de Tiempo
17.
Brain ; 134(Pt 1): 157-70, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20923787

RESUMEN

Glutaric aciduria type I, an inherited deficiency of glutaryl-coenzyme A dehydrogenase localized in the final common catabolic pathway of L-lysine, L-hydroxylysine and L-tryptophan, leads to accumulation of neurotoxic glutaric and 3-hydroxyglutaric acid, as well as non-toxic glutarylcarnitine. Most untreated patients develop irreversible brain damage during infancy that can be prevented in the majority of cases if metabolic treatment with a low L-lysine diet and L-carnitine supplementation is started in the newborn period. The biochemical effect of this treatment remains uncertain, since cerebral concentrations of neurotoxic metabolites can only be determined by invasive techniques. Therefore, we studied the biochemical effect and mechanism of metabolic treatment in glutaryl-coenzyme A dehydrogenase-deficient mice, an animal model with complete loss of glutaryl-coenzyme A dehydrogenase activity, focusing on the tissue-specific changes of neurotoxic metabolites and key enzymes of L-lysine metabolism. Here, we demonstrate that low L-lysine diet, but not L-carnitine supplementation, lowered the concentration of glutaric acid in brain, liver, kidney and serum. L-carnitine supplementation restored the free L-carnitine pool and enhanced the formation of glutarylcarnitine. The effect of low L-lysine diet was amplified by add-on therapy with L-arginine, which we propose to result from competition with L-lysine at system y(+) of the blood-brain barrier and the mitochondrial L-ornithine carriers. L-lysine can be catabolized in the mitochondrial saccharopine or the peroxisomal pipecolate pathway. We detected high activity of mitochondrial 2-aminoadipate semialdehyde synthase, the rate-limiting enzyme of the saccharopine pathway, in the liver, whereas it was absent in the brain. Since we found activity of the subsequent enzymes of L-lysine oxidation, 2-aminoadipate semialdehyde dehydrogenase, 2-aminoadipate aminotransferase and 2-oxoglutarate dehydrogenase complex as well as peroxisomal pipecolic acid oxidase in brain tissue, we postulate that the pipecolate pathway is the major route of L-lysine degradation in the brain and the saccharopine pathway is the major route in the liver. Interestingly, treatment with clofibrate decreased cerebral and hepatic concentrations of glutaric acid in glutaryl-coenzyme A dehydrogenase-deficient mice. This finding opens new therapeutic perspectives such as pharmacological stimulation of alternative L-lysine oxidation in peroxisomes. In conclusion, this study gives insight into the discrepancies between cerebral and hepatic L-lysine metabolism, provides for the first time a biochemical proof of principle for metabolic treatment in glutaric aciduria type I and suggests that further optimization of treatment could be achieved by exploitation of competition between L-lysine and L-arginine at physiological barriers and enhancement of peroxisomal L-lysine oxidation and glutaric acid breakdown.


Asunto(s)
Encéfalo/metabolismo , Lisina/metabolismo , 2-Aminoadipato-Transaminasa/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Análisis de Varianza , Animales , Arginina/metabolismo , Arginina/uso terapéutico , Encefalopatías Metabólicas/dietoterapia , Encefalopatías Metabólicas/metabolismo , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina/uso terapéutico , Catalasa/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Glutaril-CoA Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Ratones
18.
Biochim Biophys Acta ; 1802(6): 552-60, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20302929

RESUMEN

Intracerebral accumulation of neurotoxic dicarboxylic acids (DCAs) plays an important pathophysiological role in glutaric aciduria type I and methylmalonic aciduria. Therefore, we investigated the transport characteristics of accumulating DCAs - glutaric (GA), 3-hydroxyglutaric (3-OH-GA) and methylmalonic acid (MMA) - across porcine brain capillary endothelial cells (pBCEC) and human choroid plexus epithelial cells (hCPEC) representing in vitro models of the blood-brain barrier (BBB) and the choroid plexus respectively. We identified expression of organic acid transporters 1 (OAT1) and 3 (OAT3) in pBCEC on mRNA and protein level. For DCAs tested, transport from the basolateral to the apical site (i.e. efflux) was higher than influx. Efflux transport of GA, 3-OH-GA, and MMA across pBCEC was Na(+)-dependent, ATP-independent, and was inhibited by the OAT substrates para-aminohippuric acid (PAH), estrone sulfate, and taurocholate, and the OAT inhibitor probenecid. Members of the ATP-binding cassette transporter family or the organic anion transporting polypeptide family, namely MRP2, P-gp, BCRP, and OATP1B3, did not mediate transport of GA, 3-OH-GA or MMA confirming the specificity of efflux transport via OATs. In hCPEC, cellular import of GA was dependent on Na(+)-gradient, inhibited by NaCN, and unaffected by probenecid suggesting a Na(+)-dependent DCA transporter. Specific transport of GA across hCPEC, however, was not found. In conclusion, our results indicate a low but specific efflux transport for GA, 3-OH-GA, and MMA across pBCEC, an in vitro model of the BBB, via OAT1 and OAT3 but not across hCPEC, an in vitro model of the choroid plexus.


Asunto(s)
Barrera Hematoencefálica/fisiología , Encéfalo/metabolismo , Plexo Coroideo/metabolismo , Ácidos Dicarboxílicos/metabolismo , Glutaratos/orina , Ácido Metilmalónico/orina , Animales , Secuencia de Bases , Células Cultivadas , Cartilla de ADN/genética , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Técnicas In Vitro , Errores Innatos del Metabolismo/metabolismo , Metilmalonil-CoA Mutasa/deficiencia , Modelos Biológicos , Neurotoxinas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos
19.
FASEB J ; 24(8): 2938-50, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20335224

RESUMEN

Oxidative stress and increased release of reactive oxygen species (ROS) are associated with apoptosis induction. Here we report ROS-mediated induction of apoptosis by xanthohumol (XN) from hops. XN at concentrations of 1.6-25 microM induced an immediate and transient increase in superoxide anion radical (O(2)(-*)) formation in 3 human cancer cell lines (average+/-SD EC(50) of maximum O(2)(-*) induction=3.1+/-0.8 microM), murine macrophages (EC(50)=4.0+/-0.3 microM), and BPH-1 benign prostate hyperplasia cells (EC(50)=4.3+/-0.1 microM), as evidenced by the O(2)(-*)-specific indicator dihydroethidium. MitoSOX Red costaining and experiments using isolated mouse liver mitochondria (EC(50)=11.4+/-1.8 microM) confirmed mitochondria as the site of intracellular O(2)(-*) formation. Antimycin A served as positive control (EC(50)=12.4+/-0.9 microM). XN-mediated O(2)(-*) release was significantly reduced in BPH-1 rho(0) cells harboring nonfunctional mitochondria (EC(50)>25 microM) and by treatment of BPH-1 cells with vitamin C, N-acetylcysteine (NAC), or the superoxide dismutase mimetic MnTMPyP. In addition, we demonstrated a rapid 15% increase in oxidized glutathione and a dose-dependent overall thiol depletion within 6 h (IC(50)=24.3+/-11 microM). Respiratory chain complexes I-III were weakly inhibited by XN in bovine heart submitochondrial particles, but electron flux from complex I and II to complex III was significantly inhibited in BPH-1 cells, with IC(50) values of 28.1 +/- 2.4 and 24.4 +/- 5.2 microM, respectively. Within 15 min, intracellular ATP levels were significantly reduced by XN at 12.5 to 50 microM concentrations (IC(50)=26.7+/-3.7 microM). Concomitantly, XN treatment caused a rapid breakdown of the mitochondrial membrane potential and the release of cytochrome c, leading to apoptosis induction. Pre- or coincubation with 2 mM NAC and 50 microM MnTMPyP at various steps increased XN-mediated IC(50) values for cytotoxicity in BPH-1 cells from 6.7 +/- 0.2 to 12.2 +/- 0.1 and 41.4 +/- 7.6 microM, and it confirmed XN-induced O(2)(-*) as an essential trigger for apoptosis induction. In summary, we have identified mitochondria as a novel cellular target of XN action, resulting in increased O(2)(-*) production, disruption of cellular redox balance and mitochondrial integrity, and subsequent apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Flavonoides/farmacología , Mitocondrias Hepáticas/metabolismo , Neoplasias/tratamiento farmacológico , Propiofenonas/farmacología , Animales , Antineoplásicos/farmacología , Bovinos , Línea Celular Tumoral , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Flavonoides/uso terapéutico , Glutatión/metabolismo , Humanos , Neoplasias/patología , Propiofenonas/uso terapéutico , Especies Reactivas de Oxígeno , Compuestos de Sulfhidrilo/metabolismo , Superóxidos/metabolismo
20.
Chemphyschem ; 11(17): 3752-65, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21086484

RESUMEN

A series of N-4-(4'-alkoxybiphenyl)-N',N',N",N"-tetramethylguanidinium salts was synthesized with varying alkoxy chain lengths and additional N-alkyl substituents, each with a number of different counterions. X-ray crystal-structure analyses of 1b I, 1b PF(6), 2a I, and 4a I reveal bilayer structures in the solid state and, for the 1b and 1b PF(6) salts, a hydrogen-bond-type connectivity between the guanidinium N-H group and the anion is found. For the N-alkyl homologues 2a I and 4a I the anion is still oriented close to the head group, although at a larger distance. Ion pairs are present also in solution, as demonstrated by (1)H NMR: the N-H chemical shift shows a good linear correlation with the radius, and hence the hardness, of the anion. The intramolecular conformational flexibility of 1b I, 2b I, 3b I, and 4b I was studied by temperature-dependent (1)H NMR spectroscopy and discrete activation barriers were determined for rotations about each of the three C-N partial double bonds of the guanidinium core. The relative heights of the individual barriers change between the N-H and the N-alkylguanidinium salts. A fourth barrier is observed for the rotation about the N-biphenyl bond. DFT calculations of charge densities show that the positive charge resides primarily on the central carbon atom. Rotational barriers were calculated for N'-substituted 2-amino-1,3-dimethylimidazolidinium cations as models, and are in qualitatively good agreement with the NMR data. Mesomorphic properties were studied by differential-scanning calorimetry, polarizing optical microscopy, and X-ray diffraction (WAXS/SAXS). All liquid-crystalline guanidinium salts exhibit smectic A mesophases. Clearing temperatures show a linear correlation with the anionic radius. Substitution of the N-H group with methyl, ethyl, or propyl results in decreasing mesophase widths and a concomitant shrinkage of the layer spacings.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA