Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 48
Filtrer
1.
Hum Mol Genet ; 32(17): 2717-2734, 2023 08 26.
Article de Anglais | MEDLINE | ID: mdl-37369025

RÉSUMÉ

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.


Sujet(s)
Aminoacidopathies congénitales , Souris , Animaux , Aminoacidopathies congénitales/génétique , Aminoacidopathies congénitales/métabolisme , Métabolisme énergétique/génétique , Foie/métabolisme
2.
Sci Rep ; 10(1): 13584, 2020 08 12.
Article de Anglais | MEDLINE | ID: mdl-32788680

RÉSUMÉ

Warburg effect or aerobic glycolysis provides selective growth advantage to aggressive cancers. However, targeting oncogenic regulators of Warburg effect has always been challenging owing to the wide spectrum of roles of these molecules in multitude of cells. In this study, we present ADP-dependent glucokinase (ADPGK) as a novel glucose sensor and a potential onco-target in specifically high-proliferating cells in Burkitt's lymphoma (BL). Previously, we had shown ADPGK to play a major role in T-cell activation and induction of Warburg effect. We now report ADPGK knock-out Ramos BL cells display abated in vitro and in vivo tumour aggressiveness, via tumour-macrophage co-culture, migration and Zebrafish xenograft studies. We observed perturbed glycolysis and visibly reduced markers of Warburg effect in ADPGK knock-out cells, finally leading to apoptosis. We found repression of MYC proto-oncogene, and up to four-fold reduction in accumulated mutations in translocated MYC in knock-out cells, signifying a successful targeting of the malignancy. Further, the activation induced differentiation capability of knock-out cells was impaired, owing to the inability to cope up with increased energy demands. The effects amplified greatly upon stimulation-based proliferation, thus providing a novel Burkitt's lymphoma targeting mechanism originating from metabolic catastrophe induced in the cells by removal of ADPGK.


Sujet(s)
Prolifération cellulaire/génétique , Glucokinase/génétique , Tumeurs hématologiques/génétique , Danio zébré/génétique , Animaux , Apoptose/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Techniques de coculture , Techniques de knock-out de gènes , Glucokinase/métabolisme , Glycolyse/génétique , Tumeurs hématologiques/métabolisme , Tumeurs hématologiques/anatomopathologie , Humains , Cellules Jurkat , Macrophages/métabolisme , Macrophages/anatomopathologie , Proto-oncogène Mas , Lymphocytes T/métabolisme , Cellules THP-1 , Transplantation hétérologue , Danio zébré/métabolisme
3.
IUCrJ ; 7(Pt 4): 693-706, 2020 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-32695416

RÉSUMÉ

DHTKD1 is a lesser-studied E1 enzyme among the family of 2-oxoacid de-hydrogenases. In complex with E2 (di-hydro-lipo-amide succinyltransferase, DLST) and E3 (dihydrolipo-amide de-hydrogenase, DLD) components, DHTKD1 is involved in lysine and tryptophan catabolism by catalysing the oxidative de-carboxyl-ation of 2-oxoadipate (2OA) in mitochondria. Here, the 1.9 Šresolution crystal structure of human DHTKD1 is solved in complex with the thi-amine diphosphate co-factor. The structure reveals how the DHTKD1 active site is modelled upon the well characterized homologue 2-oxoglutarate (2OG) de-hydrogenase but engineered specifically to accommodate its preference for the longer substrate of 2OA over 2OG. A 4.7 Šresolution reconstruction of the human DLST catalytic core is also generated by single-particle electron microscopy, revealing a 24-mer cubic scaffold for assembling DHTKD1 and DLD protomers into a megacomplex. It is further demonstrated that missense DHTKD1 variants causing the inborn error of 2-amino-adipic and 2-oxoadipic aciduria impact on the complex formation, either directly by disrupting the interaction with DLST, or indirectly through destabilizing the DHTKD1 protein. This study provides the starting framework for developing DHTKD1 modulators to probe the intricate mitochondrial energy metabolism.

4.
Sci Rep ; 10(1): 11597, 2020 07 14.
Article de Anglais | MEDLINE | ID: mdl-32665666

RÉSUMÉ

Inspired by recent proteomic data demonstrating the upregulation of carbon and glycogen metabolism in aging human hematopoietic stem and progenitor cells (HPCs, CD34+ cells), this report addresses whether this is caused by elevated glycolysis of the HPCs on a per cell basis, or by a subpopulation that has become more glycolytic. The average glycogen content in individual CD34+ cells from older subjects (> 50 years) was 3.5 times higher and more heterogeneous compared to younger subjects (< 35 years). Representative glycolytic enzyme activities in HPCs confirmed a significant increase in glycolysis in older subjects. The HPCs from older subjects can be fractionated into three distinct subsets with high, intermediate, and low glucose uptake (GU) capacity, while the subset with a high GU capacity could scarcely be detected in younger subjects. Thus, we conclude that upregulated glycolysis in aging HPCs is caused by the expansion of a more glycolytic HPC subset. Since single-cell RNA analysis has also demonstrated that this subpopulation is linked to myeloid differentiation and increased proliferation, isolation and mechanistic characterization of this subpopulation can be utilized to elucidate specific targets for therapeutic interventions to restore the lineage balance of aging HPCs.


Sujet(s)
Carbone/métabolisme , Vieillissement de la cellule/génétique , Cellules souches hématopoïétiques/métabolisme , Cellules souches/métabolisme , Adulte , Femelle , Glycogène/métabolisme , Humains , Mâle , Adulte d'âge moyen
5.
Nat Metab ; 1(11): 1157-1167, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31742248

RÉSUMÉ

Catecholamines stimulate the first step of lipolysis by PKA-dependent release of the lipid droplet-associated protein ABHD5 from perilipin to co-activate the lipase ATGL. Here, we unmask a yet unrecognized proteolytic and cardioprotective function of ABHD5. ABHD5 acts in vivo and in vitro as a serine protease cleaving HDAC4. Through the production of an N-terminal polypeptide of HDAC4 (HDAC4-NT), ABHD5 inhibits MEF2-dependent gene expression and thereby controls glucose handling. ABHD5-deficiency leads to neutral lipid storage disease in mice. Cardiac-specific gene therapy of HDAC4-NT does not protect from intra-cardiomyocyte lipid accumulation but strikingly from heart failure, thereby challenging the concept of lipotoxicity-induced heart failure. ABHD5 levels are reduced in failing human hearts and murine transgenic ABHD5 expression protects from pressure-overload induced heart failure. These findings represent a conceptual advance by connecting lipid with glucose metabolism through HDAC4 proteolysis and enable new translational approaches to treat cardiometabolic disease.


Sujet(s)
1-Acylglycerol-3-phosphate O-acyltransferase/métabolisme , Histone deacetylases/métabolisme , Gouttelettes lipidiques , Protéines de répression/métabolisme , Cellules 3T3-L1 , Animaux , Défaillance cardiaque/prévention et contrôle , Humains , Souris , Liaison aux protéines , Protéolyse , Protéases à sérine/métabolisme
6.
Sci Rep ; 9(1): 14248, 2019 10 03.
Article de Anglais | MEDLINE | ID: mdl-31582762

RÉSUMÉ

Modulation of energy metabolism to a highly glycolytic phenotype, i.e. Warburg effect, is a common phenotype of cancer and activated immune cells allowing increased biomass-production for proliferation and cell division. Endoplasmic reticulum (ER)-localized ADP-dependent glucokinase (ADPGK) has been shown to play a critical role in T cell receptor activation-induced remodeling of energy metabolism, however the underlying mechanisms remain unclear. Therefore, we established and characterized in vitro and in vivo models for ADPGK-deficiency using Jurkat T cells and zebrafish. Upon activation, ADPGK knockout Jurkat T cells displayed increased cell death and ER stress. The increase in cell death resulted from a metabolic catastrophe and knockout cells displayed severely disturbed energy metabolism hindering induction of Warburg phenotype. ADPGK knockdown in zebrafish embryos led to short, dorsalized body axis induced by elevated apoptosis. ADPGK hypomorphic zebrafish further displayed dysfunctional glucose metabolism. In both model systems loss of ADPGK function led to defective N- and O-glycosylation. Overall, our data illustrate that ADPGK is part of a glucose sensing system in the ER modulating metabolism via regulation of N- and O-glycosylation.


Sujet(s)
Réticulum endoplasmique/métabolisme , Glucokinase/métabolisme , Glucose/métabolisme , Protéines de poisson-zèbre/métabolisme , Danio zébré/métabolisme , Animaux , Mort cellulaire , Stress du réticulum endoplasmique , Métabolisme énergétique , Glucose/analyse , Humains , Cellules Jurkat
7.
PLoS One ; 14(4): e0215162, 2019.
Article de Anglais | MEDLINE | ID: mdl-30995231

RÉSUMÉ

Dihydropteridine reductase (QDPR) catalyzes the recycling of tetrahydrobiopterin (BH4), a cofactor in dopamine, serotonin, and phenylalanine metabolism. QDPR-deficient patients develop neurological symptoms including hypokinesia, truncal hypotonia, intellectual disability and seizures. The underlying pathomechanisms are poorly understood. We established a zebrafish model for QDPR deficiency and analyzed the expression as well as function of all zebrafish QDPR homologues during embryonic development. The homologues qdpra is essential for pigmentation and phenylalanine metabolism. Qdprb1 is expressed in the proliferative zones of the optic tectum and eye. Knockdown of qdprb1 leads to up-regulation of pro-proliferative genes and increased number of phospho-histone3 positive mitotic cells. Expression of neuronal and astroglial marker genes is concomitantly decreased. Qdprb1 hypomorphic embryos develop microcephaly and reduced eye size indicating a role for qdprb1 in the transition from cell proliferation to differentiation. Glutamine accumulation biochemically accompanies the developmental changes. Our findings provide novel insights into the neuropathogenesis of QDPR deficiency.


Sujet(s)
Prolifération cellulaire/génétique , Glutamine , Mélanines , Névroglie/métabolisme , Phénylcétonuries , Danio zébré , Animaux , Dihydropteridine reductase/génétique , Modèles animaux de maladie humaine , Techniques de knock-down de gènes , Glutamine/génétique , Glutamine/métabolisme , Humains , Mélanines/biosynthèse , Mélanines/génétique , Phénylcétonuries/génétique , Phénylcétonuries/métabolisme , Danio zébré/embryologie , Danio zébré/génétique , Protéines de poisson-zèbre/génétique
8.
Circulation ; 137(24): 2592-2608, 2018 06 12.
Article de Anglais | MEDLINE | ID: mdl-29353241

RÉSUMÉ

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.


Sujet(s)
Endothélium vasculaire/métabolisme , Acides gras/métabolisme , Myocarde/métabolisme , Récepteurs Notch/métabolisme , Transduction du signal , Remodelage vasculaire , Protéines adaptatrices de la transduction du signal , Angiopoïétines/génétique , Angiopoïétines/métabolisme , Animaux , Antigènes CD36/génétique , Antigènes CD36/métabolisme , Protéines de liaison au calcium , Endothélium vasculaire/cytologie , Protéines de liaison aux acides gras/génétique , Protéines de liaison aux acides gras/métabolisme , Acides gras/génétique , Glucose/génétique , Glucose/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Souris transgéniques , Myocytes cardiaques/métabolisme , Néovascularisation physiologique , Récepteurs Notch/génétique , Ribosomal Protein S6 Kinases/génétique , Ribosomal Protein S6 Kinases/métabolisme , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/métabolisme
9.
Nat Med ; 24(1): 62-72, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29227474

RÉSUMÉ

The stress-responsive epigenetic repressor histone deacetylase 4 (HDAC4) regulates cardiac gene expression. Here we show that the levels of an N-terminal proteolytically derived fragment of HDAC4, termed HDAC4-NT, are lower in failing mouse hearts than in healthy control hearts. Virus-mediated transfer of the portion of the Hdac4 gene encoding HDAC4-NT into the mouse myocardium protected the heart from remodeling and failure; this was associated with decreased expression of Nr4a1, which encodes a nuclear orphan receptor, and decreased NR4A1-dependent activation of the hexosamine biosynthetic pathway (HBP). Conversely, exercise enhanced HDAC4-NT levels, and mice with a cardiomyocyte-specific deletion of Hdac4 show reduced exercise capacity, which was characterized by cardiac fatigue and increased expression of Nr4a1. Mechanistically, we found that NR4A1 negatively regulated contractile function in a manner that depended on the HBP and the calcium sensor STIM1. Our work describes a new regulatory axis in which epigenetic regulation of a metabolic pathway affects calcium handling. Activation of this axis during intermittent physiological stress promotes cardiac function, whereas its impairment in sustained pathological cardiac stress leads to heart failure.


Sujet(s)
Défaillance cardiaque/métabolisme , Hexosamine/biosynthèse , Histone deacetylases/métabolisme , Contraction myocardique , Animaux , Épigenèse génétique , Techniques de transfert de gènes , Défaillance cardiaque/génétique , Histone deacetylases/génétique , Souris , Souris knockout , Myocarde/enzymologie , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Conditionnement physique d'animal , Protéolyse , Molécule-1 d'interaction stromale/métabolisme
10.
J Inherit Metab Dis ; 41(1): 91-99, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-29098534

RÉSUMÉ

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.


Sujet(s)
Aminoacidopathies congénitales/métabolisme , Astrocytes/effets des médicaments et des substances chimiques , Encéphalopathies métaboliques/métabolisme , Antienzymes/toxicité , Glutamate dehydrogenase/antagonistes et inhibiteurs , Glutamine/métabolisme , Glutarates/toxicité , Glutaryl-CoA dehydrogenase/déficit , Aminoacidopathies congénitales/anatomopathologie , Animaux , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Encéphalopathies métaboliques/anatomopathologie , Mort cellulaire/effets des médicaments et des substances chimiques , Hypoxie cellulaire , Lignée cellulaire , Glutamate dehydrogenase/métabolisme , Glutamine/pharmacologie , Glutaryl-CoA dehydrogenase/métabolisme , Rats
11.
J Mol Med (Berl) ; 95(8): 851-860, 2017 08.
Article de Anglais | MEDLINE | ID: mdl-28585096

RÉSUMÉ

Expression of the hepatic peptide hormone hepcidin responds to iron levels via BMP/SMAD signaling, to inflammatory cues via JAK/STAT signaling, to the nutrient-sensing mTOR pathway, as well as to proliferative signals and gluconeogenesis. Here, we asked the question whether hepcidin expression is altered by metabolites generated by intermediary metabolism. To identify such metabolites, we took advantage of a comprehensive RNAi screen, which revealed effectors involved in citrate metabolism. We show that the inhibition of citrate-consuming enzymes increases hepcidin mRNA expression in primary murine hepatocytes. Consistently, citrate treatment of primary murine hepatocytes or intravenous injection of citrate in mice increases cellular citrate concentrations and hepcidin expression. We further demonstrate that the hepcidin response to citrate involves the SMAD signaling pathway. These results reveal links between iron homeostasis and energy metabolism that may help to explain why iron levels are frequently altered in metabolic disorders. KEY MESSAGES: • Elevated citrate levels increase hepcidin mRNA expression in primary hepatocytes. • Citrate treatment in primary hepatocytes activates hepcidin expression. • Intravenous injection of citrate in mice increases hepcidin mRNA levels. • The hepcidin response to citrate involves the BMP/SMAD signaling pathway.


Sujet(s)
Acide citrique/métabolisme , Métabolisme énergétique , Hépatocytes/métabolisme , Hepcidines/génétique , Aconitate hydratase/génétique , Animaux , Cellules cultivées , Hepcidines/sang , Homéostasie , Fer/métabolisme , Protéine-1 de régulation du fer/génétique , Mâle , Souris de lignée C57BL , Interférence par ARN , ARN messager/métabolisme , Transduction du signal , Protéines Smad/métabolisme
12.
Biochim Biophys Acta Mol Basis Dis ; 1863(9): 2220-2228, 2017 09.
Article de Anglais | MEDLINE | ID: mdl-28545977

RÉSUMÉ

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.


Sujet(s)
Aminoacidopathies congénitales/métabolisme , Encéphalopathies métaboliques/métabolisme , Encéphale/métabolisme , Glutarates/métabolisme , Glutaryl-CoA dehydrogenase/déficit , Cetone oxidoreductases/déficit , Foie/métabolisme , Lysine/métabolisme , Aminoacidopathies congénitales/génétique , Aminoacidopathies congénitales/thérapie , Animaux , Encéphale/anatomopathologie , Encéphalopathies métaboliques/génétique , Encéphalopathies métaboliques/thérapie , Modèles animaux de maladie humaine , Glutaryl-CoA dehydrogenase/génétique , Glutaryl-CoA dehydrogenase/métabolisme , Cetone oxidoreductases/métabolisme , Foie/anatomopathologie , Souris , Souris knockout
13.
Am J Hum Genet ; 99(2): 414-22, 2016 Aug 04.
Article de Anglais | MEDLINE | ID: mdl-27426735

RÉSUMÉ

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.


Sujet(s)
Allèles , Retard de croissance intra-utérin/génétique , Déficience intellectuelle/génétique , Isoleucine-tRNA ligase/génétique , Maladies du foie/congénital , Maladies du foie/génétique , Hypotonie musculaire/congénital , Hypotonie musculaire/génétique , Mutation , Adolescent , Animaux , Enfant , Enfant d'âge préscolaire , Compléments alimentaires , Stéatose hépatique/génétique , Femelle , Fibrose/génétique , Humains , Nourrisson , Nouveau-né , Isoleucine-tRNA ligase/déficit , Défaillance hépatique/génétique , Mâle , Syndrome , Danio zébré/génétique , Zinc/administration et posologie , Zinc/déficit , Zinc/usage thérapeutique
14.
J Inherit Metab Dis ; 39(5): 625-632, 2016 09.
Article de Anglais | MEDLINE | ID: mdl-27139199

RÉSUMÉ

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.


Sujet(s)
Aminoacidopathies congénitales/étiologie , Aminoacidopathies congénitales/génétique , Mutation/génétique , Oxidoreductases acting on CH-NH group donors/déficit , Enfant , Enfant d'âge préscolaire , Femelle , Allemagne , Hétérozygote , Homozygote , Humains , Hydroxyproline/génétique , Nourrisson , Nouveau-né , Mâle , Leucinose/étiologie , Leucinose/génétique , Dépistage néonatal/méthodes , Oxidoreductases acting on CH-NH group donors/génétique , Phénotype , Prévalence , Proline dehydrogenase/génétique
15.
Nat Commun ; 7: 10764, 2016 Mar 07.
Article de Anglais | MEDLINE | ID: mdl-26948869

RÉSUMÉ

The high-mobility group box 1 (HMGB1) protein has a central role in immunological antitumour defense. Here we show that natural killer cell-derived HMGB1 directly eliminates cancer cells by triggering metabolic cell death. HMGB1 allosterically inhibits the tetrameric pyruvate kinase isoform M2, thus blocking glucose-driven aerobic respiration. This results in a rapid metabolic shift forcing cells to rely solely on glycolysis for the maintenance of energy production. Cancer cells can acquire resistance to HMGB1 by increasing glycolysis using the dimeric form of PKM2, and employing glutaminolysis. Consistently, we observe an increase in the expression of a key enzyme of glutaminolysis, malic enzyme 1, in advanced colon cancer. Moreover, pharmaceutical inhibition of glutaminolysis sensitizes tumour cells to HMGB1 providing a basis for a therapeutic strategy for treating cancer.


Sujet(s)
Tumeurs du côlon/métabolisme , Tumeurs du côlon/physiopathologie , Protéine HMGB1/métabolisme , Protéines de transport/génétique , Protéines de transport/métabolisme , Mort cellulaire , Lignée cellulaire tumorale , Respiration cellulaire , Tumeurs du côlon/enzymologie , Tumeurs du côlon/génétique , Glucose/métabolisme , Glycolyse , Protéine HMGB1/génétique , Humains , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Hormones thyroïdiennes/génétique , Hormones thyroïdiennes/métabolisme ,
16.
Aging (Albany NY) ; 7(11): 911-27, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26546739

RÉSUMÉ

The TERT gene encodes for the reverse transcriptase activity of the telomerase complex and mutations in TERT can lead to dysfunctional telomerase activity resulting in diseases such as dyskeratosis congenita (DKC). Here, we describe a novel TERT mutation at position T1129P leading to DKC with progressive bone marrow (BM) failure in homozygous members of a consanguineous family. BM hematopoietic stem cells (HSCs) of an affected family member were 300-fold reduced associated with a significantly impaired colony forming capacity in vitro and impaired repopulation activity in mouse xenografts. Recent data in yeast suggested improved cellular checkpoint controls by mTOR inhibition preventing cells with short telomeres or DNA damage from dividing. To evaluate a potential therapeutic option for the patient, we treated her primary skin fibroblasts and BM HSCs with the mTOR inhibitor rapamycin. This led to prolonged survival and decreased levels of senescence in T1129P mutant fibroblasts. In contrast, the impaired HSC function could not be improved by mTOR inhibition, as colony forming capacity and multilineage engraftment potential in xenotransplanted mice remained severely impaired. Thus, rapamycin treatment did not rescue the compromised stem cell function of TERTT1129P mutant patient HSCs and outlines limitations of a potential DKC therapy based on rapamycin.


Sujet(s)
Antigènes CD34/analyse , Vieillissement de la cellule , Dyskératose congénitale/génétique , Cellules souches hématopoïétiques/physiologie , Mutation , Sirolimus/pharmacologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Telomerase/génétique , Animaux , Femelle , Cellules HeLa , Transplantation de cellules souches hématopoïétiques , Humains , Souris , Télomère
17.
RNA ; 21(11): 1873-84, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26370583

RÉSUMÉ

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.


Sujet(s)
Protéines mitochondriales/génétique , Biosynthèse des protéines/génétique , Protein-Serine-Threonine Kinases/génétique , Protéines de liaison à l'ARN/génétique , ARN/génétique , Lignée cellulaire , Respiration cellulaire/génétique , Cellules HEK293 , Humains , Mitochondries/génétique , NADH dehydrogenase/génétique , ARN messager/génétique , ARN mitochondrial , ARN ribosomique 16S/génétique
18.
PLoS One ; 10(6): e0128770, 2015.
Article de Anglais | MEDLINE | ID: mdl-26086473

RÉSUMÉ

Maleic acid (MA) has been shown to induce Fanconi syndrome via disturbance of renal energy homeostasis, though the underlying pathomechanism is still under debate. Our study aimed to examine the pathomechanism underlying maleic acid-induced nephrotoxicity. Methylmalonic acid (MMA) is structurally similar to MA and accumulates in patients affected with methymalonic aciduria, a defect in the degradation of branched-chain amino acids, odd-chain fatty acids and cholesterol, which is associated with the development of tubulointerstitial nephritis resulting in chronic renal failure. We therefore used MMA application as a control experiment in our study and stressed hPTECs with MA and MMA to further validate the specificity of our findings. MMA did not show any toxic effects on proximal tubule cells, whereas maleic acid induced concentration-dependent and time-dependent cell death shown by increased lactate dehydrogenase release as well as ethidium homodimer and calcein acetoxymethyl ester staining. The toxic effect of MA was blocked by administration of single amino acids, in particular L-alanine and L-glutamate. MA application further resulted in severe impairment of cellular energy homeostasis on the level of glycolysis, respiratory chain, and citric acid cycle resulting in ATP depletion. As underlying mechanism we could identify disturbance of calcium homeostasis. MA toxicity was critically dependent on calcium levels in culture medium and blocked by the extra- and intracellular calcium chelators EGTA and BAPTA-AM respectively. Moreover, MA-induced cell death was associated with activation of calcium-dependent calpain proteases. In summary, our study shows a comprehensive pathomechanistic concept for MA-induced dysfunction and damage of human proximal tubule cells.


Sujet(s)
Calcium/métabolisme , Métabolisme énergétique/effets des médicaments et des substances chimiques , Homéostasie/effets des médicaments et des substances chimiques , Maléates/pharmacologie , Acide méthyl-malonique/pharmacologie , Calcium/physiologie , Cellules cultivées , Relation dose-effet des médicaments , Humains , Défaillance rénale chronique/induit chimiquement , Tubules contournés proximaux/cytologie , Tubules contournés proximaux/effets des médicaments et des substances chimiques , L-Lactate dehydrogenase/métabolisme , Consommation d'oxygène/effets des médicaments et des substances chimiques , Relation structure-activité
19.
Biochim Biophys Acta ; 1852(5): 768-77, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25558815

RÉSUMÉ

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.


Sujet(s)
Aminoacidopathies congénitales/génétique , Encéphalopathies métaboliques/génétique , Prédisposition génétique à une maladie/génétique , Glutaryl-CoA dehydrogenase/déficit , Glutaryl-CoA dehydrogenase/génétique , Lysine/administration et posologie , Aconitate hydratase/métabolisme , Aminoacidopathies congénitales/étiologie , Aminoacidopathies congénitales/métabolisme , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Encéphale/anatomopathologie , Encéphalopathies métaboliques/étiologie , Encéphalopathies métaboliques/métabolisme , Régime alimentaire , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Métabolisme énergétique/effets des médicaments et des substances chimiques , Femelle , Prédisposition génétique à une maladie/étiologie , Glutarates/métabolisme , Glutaryl-CoA dehydrogenase/métabolisme , Glyceraldehyde 3-phosphate dehydrogenases/métabolisme , Ketoglutarate dehydrogenase complex/métabolisme , Lysine/effets indésirables , Mâle , Souris de souche-129 , Souris de lignée C57BL , Souris knockout , Facteurs de risque , Facteurs sexuels , Spectrophotométrie
20.
J Inherit Metab Dis ; 38(2): 265-72, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25214427

RÉSUMÉ

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.


Sujet(s)
Aminoacidopathies congénitales/enzymologie , Aminoacidopathies congénitales/génétique , Encéphalopathies métaboliques/enzymologie , Encéphalopathies métaboliques/génétique , Encéphale/enzymologie , Glutaryl-CoA dehydrogenase/déficit , Glutaryl-CoA dehydrogenase/génétique , Lysine/métabolisme , Acides pipécoliques/métabolisme , Animaux , Désamination , Modèles animaux de maladie humaine , Prédisposition génétique à une maladie , Foie/enzymologie , Lysine/analogues et dérivés , Souris knockout , Oxydoréduction , Oxidoreductases acting on CH-NH group donors/métabolisme , Péroxysomes/enzymologie , Phénotype
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...