Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Metab ; 1(11): 1157-1167, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31742248

RESUMO

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.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/metabolismo , Histona Desacetilases/metabolismo , Gotículas Lipídicas , Proteínas Repressoras/metabolismo , Células 3T3-L1 , Animais , Insuficiência Cardíaca/prevenção & controle , Humanos , Camundongos , Ligação Proteica , Proteólise , Serina Proteases/metabolismo
2.
J Mol Med (Berl) ; 95(8): 851-860, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28585096

RESUMO

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.


Assuntos
Ácido Cítrico/metabolismo , Metabolismo Energético , Hepatócitos/metabolismo , Hepcidinas/genética , Aconitato Hidratase/genética , Animais , Células Cultivadas , Hepcidinas/sangue , Homeostase , Ferro/metabolismo , Proteína 1 Reguladora do Ferro/genética , Masculino , Camundongos Endogâmicos C57BL , Interferência de RNA , RNA Mensageiro/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo
3.
Nat Commun ; 7: 10764, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26948869

RESUMO

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.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/fisiopatologia , Proteína HMGB1/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Morte Celular , Linhagem Celular Tumoral , Respiração Celular , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Glucose/metabolismo , Glicólise , Proteína HMGB1/genética , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Hormônios Tireóideos/genética , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
4.
Aging (Albany NY) ; 7(11): 911-27, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26546739

RESUMO

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.


Assuntos
Antígenos CD34/análise , Senescência Celular , Disceratose Congênita/genética , Células-Tronco Hematopoéticas/fisiologia , Mutação , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Telomerase/genética , Animais , Feminino , Células HeLa , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Telômero
5.
J Inherit Metab Dis ; 36(4): 635-44, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23512157

RESUMO

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.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/metabolismo , Encefalopatias Metabólicas Congênitas/metabolismo , Encéfalo/metabolismo , Erros Inatos do Metabolismo dos Aminoácidos/patologia , Animais , Encéfalo/patologia , Encefalopatias Metabólicas Congênitas/patologia , Metabolismo Energético , Humanos
6.
Biochim Biophys Acta ; 1812(12): 1607-15, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21920437

RESUMO

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.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Colesterol/metabolismo , Degeneração Hepatolenticular/metabolismo , Aconitato Hidratase/metabolismo , Alanina/metabolismo , Animais , Encéfalo/enzimologia , Encéfalo/metabolismo , Carnitina/análogos & derivados , Carnitina/sangue , Carnitina/metabolismo , Colesterol/sangue , Cobre/metabolismo , ATPases Transportadoras de Cobre , Modelos Animais de Doenças , Transporte de Elétrons , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Técnicas de Inativação de Genes , Glutationa/metabolismo , Degeneração Hepatolenticular/sangue , Humanos , Fígado/enzimologia , Fígado/metabolismo , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Mitocôndrias/metabolismo , Estresse Oxidativo , Superóxido Dismutase/metabolismo
7.
Cell Metab ; 12(2): 194-201, 2010 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-20674864

RESUMO

Mitochondria supply cells with ATP, heme, and iron sulfur clusters (ISC), and mitochondrial energy metabolism involves both heme- and ISC-dependent enzymes. Here, we show that mitochondrial iron supply and function require iron regulatory proteins (IRP), cytosolic RNA-binding proteins that control mRNA translation and stability. Mice lacking both IRP1 and IRP2 in their hepatocytes suffer from mitochondrial iron deficiency and dysfunction associated with alterations of the ISC and heme biosynthetic pathways, leading to liver failure and death. These results uncover a major role of the IRPs in cell biology: to ensure adequate iron supply to the mitochondrion for proper function of this critical organelle.


Assuntos
Proteínas Reguladoras de Ferro/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Animais , Metabolismo Energético , Heme/biossíntese , Proteína 1 Reguladora do Ferro/deficiência , Proteína 1 Reguladora do Ferro/genética , Proteína 1 Reguladora do Ferro/metabolismo , Proteína 2 Reguladora do Ferro/deficiência , Proteína 2 Reguladora do Ferro/genética , Proteína 2 Reguladora do Ferro/metabolismo , Proteínas Reguladoras de Ferro/deficiência , Proteínas Reguladoras de Ferro/genética , Falência Hepática/etiologia , Falência Hepática/metabolismo , Falência Hepática/patologia , Camundongos , Camundongos Transgênicos
8.
Biochim Biophys Acta ; 1802(6): 552-60, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20302929

RESUMO

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.


Assuntos
Barreira Hematoencefálica/fisiologia , Encéfalo/metabolismo , Plexo Corióideo/metabolismo , Ácidos Dicarboxílicos/metabolismo , Glutaratos/urina , Ácido Metilmalônico/urina , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA/genética , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Glutaril-CoA Desidrogenase/deficiência , Humanos , Técnicas In Vitro , Erros Inatos do Metabolismo/metabolismo , Metilmalonil-CoA Mutase/deficiência , Modelos Biológicos , Neurotoxinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Suínos
9.
FASEB J ; 24(8): 2938-50, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20335224

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Flavonoides/farmacologia , Mitocôndrias Hepáticas/metabolismo , Neoplasias/tratamento farmacológico , Propiofenonas/farmacologia , Animais , Antineoplásicos/farmacologia , Bovinos , Linhagem Celular Tumoral , Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Flavonoides/uso terapêutico , Glutationa/metabolismo , Humanos , Neoplasias/patologia , Propiofenonas/uso terapêutico , Espécies Reativas de Oxigênio , Compostos de Sulfidrila/metabolismo , Superóxidos/metabolismo
10.
Biochem J ; 398(1): 107-12, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16686602

RESUMO

Mitochondrial dysfunction during acute metabolic crises is considered an important pathomechanism in inherited disorders of propionate metabolism, i.e. propionic and methylmalonic acidurias. Biochemically, these disorders are characterized by accumulation of propionyl-CoA and metabolites of alternative propionate oxidation. In the present study, we demonstrate uncompetitive inhibition of PDHc (pyruvate dehydrogenase complex) by propionyl-CoA in purified porcine enzyme and in submitochondrial particles from bovine heart being in the same range as the inhibition induced by acetyl-CoA, the physiological product and known inhibitor of PDHc. Evaluation of similar monocarboxylic CoA esters showed a chain-length specificity for PDHc inhibition. In contrast with CoA esters, non-esterified fatty acids did not inhibit PDHc activity. In addition to PDHc inhibition, analysis of respiratory chain and tricarboxylic acid cycle enzymes also revealed an inhibition by propionyl-CoA on respiratory chain complex III and alpha-ketoglutarate dehydrogenase complex. To test whether impairment of mitochondrial energy metabolism is involved in the pathogenesis of propionic aciduria, we performed a thorough bioenergetic analysis in muscle biopsy specimens of two patients. In line with the in vitro results, oxidative phosphorylation was severely compromised in both patients. Furthermore, expression of respiratory chain complexes I-IV and the amount of mitochondrial DNA were strongly decreased, and ultrastructural mitochondrial abnormalities were found, highlighting severe mitochondrial dysfunction. In conclusion, our results favour the hypothesis that toxic metabolites, in particular propionyl-CoA, are involved in the pathogenesis of inherited disorders of propionate metabolism, sharing mechanistic similarities with propionate toxicity in micro-organisms.


Assuntos
Erros Inatos do Metabolismo dos Aminoácidos/complicações , Doenças Mitocondriais/etiologia , Doenças Mitocondriais/fisiopatologia , Propionatos/metabolismo , Toxinas Biológicas/metabolismo , Acetilcoenzima A/farmacologia , Acil Coenzima A/farmacologia , Animais , Bovinos , Metabolismo Energético/efeitos dos fármacos , Ácidos Graxos/farmacologia , Feminino , Fibroblastos/enzimologia , Humanos , Recém-Nascido , Masculino , Doenças Mitocondriais/metabolismo , Fosforilação Oxidativa , Propionatos/toxicidade , Complexo Piruvato Desidrogenase/antagonistas & inibidores , Músculo Quadríceps/ultraestrutura , Pele/enzimologia , Suínos , Toxinas Biológicas/toxicidade
11.
J Biol Chem ; 280(23): 21830-6, 2005 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-15840571

RESUMO

Inherited deficiency of glutaryl-CoA dehydrogenase results in an accumulation of glutaryl-CoA, glutaric, and 3-hydroxyglutaric acids. If untreated, most patients suffer an acute encephalopathic crisis and, subsequently, acute striatal damage being precipitated by febrile infectious diseases during a vulnerable period of brain development (age 3 and 36 months). It has been suggested before that some of these organic acids may induce excitotoxic cell damage, however, the relevance of bioenergetic impairment is not yet understood. The major aim of our study was to investigate respiratory chain, tricarboxylic acid cycle, and fatty acid oxidation in this disease using purified single enzymes and tissue homogenates from Gcdh-deficient and wild-type mice. In purified enzymes, glutaryl-CoA but not glutaric or 3-hydroxyglutaric induced an uncompetitive inhibition of alpha-ketoglutarate dehydrogenase complex activity. Notably, reduced activity of alpha-ketoglutarate dehydrogenase activity has recently been demonstrated in other neurodegenerative diseases, such as Alzheimer, Parkinson, and Huntington diseases. In contrast to alpha-ketoglutarate dehydrogenase complex, no direct inhibition of glutaryl-CoA, glutaric acid, and 3-hydroxyglutaric acid was found in other enzymes tested. In Gcdh-deficient mice, respiratory chain and tricarboxylic acid activities remained widely unaffected, virtually excluding regulatory changes in these enzymes. However, hepatic activity of very long-chain acyl-CoA dehydrogenase was decreased and concentrations of long-chain acylcarnitines increased in the bile of these mice, which suggested disturbed oxidation of long-chain fatty acids. In conclusion, our results demonstrate that bioenergetic impairment may play an important role in the pathomechanisms underlying neurodegenerative changes in glutaryl-CoA dehydrogenase deficiency.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH/deficiência , Aconitato Hidratase/metabolismo , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Bovinos , Ciclo do Ácido Cítrico , Di-Hidrolipoamida Desidrogenase/metabolismo , Relação Dose-Resposta a Droga , Ácidos Graxos/metabolismo , Glutaril-CoA Desidrogenase , Glutationa/metabolismo , Isocitrato Desidrogenase/metabolismo , Complexo Cetoglutarato Desidrogenase/metabolismo , Cetona Oxirredutases/metabolismo , Cinética , Fígado/metabolismo , Camundongos , Camundongos Transgênicos , Miocárdio/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Oxigênio/metabolismo , Espectrofotometria
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA