Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Blood ; 134(22): 1983-1995, 2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-31570489

RESUMO

Host cell competition is a major barrier to engraftment after in utero hematopoietic cell transplantation (IUHCT). Here we describe a cell-engineering strategy using glycogen synthase kinase-3 (GSK3) inhibitor-loaded nanoparticles conjugated to the surface of donor hematopoietic cells to enhance their proliferation kinetics and ability to compete against their fetal host equivalents. With this approach, we achieved remarkable levels of stable, long-term hematopoietic engraftment for up to 24 weeks post-IUHCT. We also show that the salutary effects of the nanoparticle-released GSK3 inhibitor are specific to donor progenitor/stem cells and achieved by a pseudoautocrine mechanism. These results establish that IUHCT of hematopoietic cells decorated with GSK3 inhibitor-loaded nanoparticles can produce therapeutic levels of long-term engraftment and could therefore allow single-step prenatal treatment of congenital hematological disorders.


Assuntos
Comunicação Autócrina , Engenharia Celular , Inibidores Enzimáticos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Sobrevivência de Enxerto/efeitos dos fármacos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Nanopartículas/química , Animais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C
2.
Sci Transl Med ; 9(416)2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29141885

RESUMO

Gut dysbiosis during inflammatory bowel disease involves alterations in the gut microbiota associated with inflammation of the host gut. We used a combination of shotgun metagenomic sequencing and metabolomics to analyze fecal samples from pediatric patients with Crohn's disease and found an association between disease severity, gut dysbiosis, and bacterial production of free amino acids. Nitrogen flux studies using 15N in mice showed that activity of bacterial urease, an enzyme that releases ammonia by hydrolysis of host urea, led to the transfer of murine host-derived nitrogen to the gut microbiota where it was used for amino acid synthesis. Inoculation of a conventional murine host (pretreated with antibiotics and polyethylene glycol) with commensal Escherichia coli engineered to express urease led to dysbiosis of the gut microbiota, resulting in a predominance of Proteobacteria species. This was associated with a worsening of immune-mediated colitis in these animals. A potential role for altered urease expression and nitrogen flux in the development of gut dysbiosis suggests that bacterial urease may be a potential therapeutic target for inflammatory bowel diseases.


Assuntos
Proteínas de Bactérias/metabolismo , Doença de Crohn/metabolismo , Doença de Crohn/microbiologia , Disbiose/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Urease/metabolismo , Animais , Humanos , Camundongos
3.
Sci Signal ; 8(384): ra68, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26152695

RESUMO

Nitric oxide (NO) is a signaling intermediate during glutamatergic neurotransmission in the central nervous system (CNS). NO signaling is in part accomplished through cysteine S-nitrosylation, a posttranslational modification by which NO regulates protein function and signaling. In our investigation of the protein targets and functional impact of S-nitrosylation in the CNS under physiological conditions, we identified 269 S-nitrosocysteine residues in 136 proteins in the wild-type mouse brain. The number of sites was significantly reduced in the brains of mice lacking endothelial nitric oxide synthase (eNOS(-/-)) or neuronal nitric oxide synthase (nNOS(-/-)). In particular, nNOS(-/-) animals showed decreased S-nitrosylation of proteins that participate in the glutamate/glutamine cycle, a metabolic process by which synaptic glutamate is recycled or oxidized to provide energy. (15)N-glutamine-based metabolomic profiling and enzymatic activity assays indicated that brain extracts from nNOS(-/-) mice converted less glutamate to glutamine and oxidized more glutamate than those from mice of the other genotypes. GLT1 [also known as EAAT2 (excitatory amino acid transporter 2)], a glutamate transporter in astrocytes, was S-nitrosylated at Cys(373) and Cys(561) in wild-type and eNOS(-/-) mice, but not in nNOS(-/-) mice. A form of rat GLT1 that could not be S-nitrosylated at the equivalent sites had increased glutamate uptake compared to wild-type GLT1 in cells exposed to an S-nitrosylating agent. Thus, NO modulates glutamatergic neurotransmission through the selective, nNOS-dependent S-nitrosylation of proteins that govern glutamate transport and metabolism.


Assuntos
Encéfalo/metabolismo , Cisteína/metabolismo , Ácido Glutâmico/metabolismo , Óxido Nítrico/metabolismo , Sequência de Aminoácidos , Animais , Western Blotting , Cromatografia Líquida , Cisteína/análogos & derivados , Cisteína/genética , Transportador 2 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Mutação , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Ratos , S-Nitrosotióis/metabolismo , Espectrometria de Massas em Tandem
4.
J Clin Invest ; 125(7): 2841-50, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26098218

RESUMO

Increasing evidence indicates that the gut microbiota can be altered to ameliorate or prevent disease states, and engineering the gut microbiota to therapeutically modulate host metabolism is an emerging goal of microbiome research. In the intestine, bacterial urease converts host-derived urea to ammonia and carbon dioxide, contributing to hyperammonemia-associated neurotoxicity and encephalopathy in patients with liver disease. Here, we engineered murine gut microbiota to reduce urease activity. Animals were depleted of their preexisting gut microbiota and then inoculated with altered Schaedler flora (ASF), a defined consortium of 8 bacteria with minimal urease gene content. This protocol resulted in establishment of a persistent new community that promoted a long-term reduction in fecal urease activity and ammonia production. Moreover, in a murine model of hepatic injury, ASF transplantation was associated with decreased morbidity and mortality. These results provide proof of concept that inoculation of a prepared host with a defined gut microbiota can lead to durable metabolic changes with therapeutic utility.


Assuntos
Terapia Biológica/métodos , Sistema Digestório/microbiologia , Hiperamonemia/microbiologia , Hiperamonemia/terapia , Microbiota , Amônia/metabolismo , Animais , Bactérias/enzimologia , Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bioengenharia , Doença Hepática Induzida por Substâncias e Drogas/terapia , Sistema Digestório/metabolismo , Modelos Animais de Doenças , Fezes/microbiologia , Feminino , Genes Bacterianos , Hiperamonemia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microbiota/fisiologia , Fatores de Tempo , Urease/genética , Urease/metabolismo
5.
J Pediatr ; 165(2): 401-403.e3, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24880889

RESUMO

Identical studies using stable isotopes were performed before and after a 3-day trial of oral N-carbamyl-l-glutamate (NCG) in 5 subjects with late-onset carbamyl phosphate synthetase deficiency. NCG augmented ureagenesis and decreased plasma ammonia in 4 of 5 subjects. There was marked improvement in nitrogen metabolism with long-term NCG administration in 1 subject.


Assuntos
Doença da Deficiência da Carbamoil-Fosfato Sintase I/tratamento farmacológico , Glutamatos/uso terapêutico , Glutamina/sangue , Ureia/metabolismo , Adolescente , Adulto , Amônia/sangue , Doença da Deficiência da Carbamoil-Fosfato Sintase I/sangue , Criança , Pré-Escolar , Feminino , Humanos , Modelos Lineares , Masculino , Espectrometria de Massas , Resultado do Tratamento , Adulto Jovem
6.
J Biol Chem ; 289(14): 9710-29, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24523404

RESUMO

Agmatine (AGM), a product of arginine decarboxylation, influences multiple physiologic and metabolic functions. However, the mechanism(s) of action, the impact on whole body gene expression and metabolic pathways, and the potential benefits and risks of long term AGM consumption are still a mystery. Here, we scrutinized the impact of AGM on whole body metabolic profiling and gene expression and assessed a plausible mechanism(s) of AGM action. Studies were performed in rats fed a high fat diet or standard chow. AGM was added to drinking water for 4 or 8 weeks. We used (13)C or (15)N tracers to assess metabolic reactions and fluxes and real time quantitative PCR to determine gene expression. The results demonstrate that AGM elevated the synthesis and tissue level of cAMP. Subsequently, AGM had a widespread impact on gene expression and metabolic profiling including (a) activation of peroxisomal proliferator-activated receptor-α and its coactivator, PGC1α, and (b) increased expression of peroxisomal proliferator-activated receptor-γ and genes regulating thermogenesis, gluconeogenesis, and carnitine biosynthesis and transport. The changes in gene expression were coupled with improved tissue and systemic levels of carnitine and short chain acylcarnitine, increased ß-oxidation but diminished incomplete fatty acid oxidation, decreased fat but increased protein mass, and increased hepatic ureagenesis and gluconeogenesis but decreased glycolysis. These metabolic changes were coupled with reduced weight gain and a curtailment of the hormonal and metabolic derangements associated with high fat diet-induced obesity. The findings suggest that AGM elevated the synthesis and levels of cAMP, thereby mimicking the effects of caloric restriction with respect to metabolic reprogramming.


Assuntos
Agmatina/farmacologia , AMP Cíclico/metabolismo , Ácidos Graxos/metabolismo , Gluconeogênese/efeitos dos fármacos , Fígado/metabolismo , Obesidade/tratamento farmacológico , Agmatina/farmacocinética , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Carnitina/análogos & derivados , Carnitina/metabolismo , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Metaboloma , Obesidade/induzido quimicamente , Obesidade/metabolismo , Oxirredução/efeitos dos fármacos , PPAR gama/biossíntese , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Fatores de Transcrição/biossíntese
7.
J Biol Chem ; 288(6): 3938-51, 2013 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-23266825

RESUMO

Paracrine signaling between pancreatic islet ß-cells and α-cells has been proposed to play a role in regulating glucagon responses to elevated glucose and hypoglycemia. To examine this possibility in human islets, we used a metabolomic approach to trace the responses of amino acids and other potential neurotransmitters to stimulation with [U-(13)C]glucose in both normal individuals and type 2 diabetics. Islets from type 2 diabetics uniformly showed decreased glucose stimulation of insulin secretion and respiratory rate but demonstrated two different patterns of glucagon responses to glucose: one group responded normally to suppression of glucagon by glucose, but the second group was non-responsive. The non-responsive group showed evidence of suppressed islet GABA levels and of GABA shunt activity. In further studies with normal human islets, we found that γ-hydroxybutyrate (GHB), a potent inhibitory neurotransmitter, is generated in ß-cells by an extension of the GABA shunt during glucose stimulation and interacts with α-cell GHB receptors, thus mediating the suppressive effect of glucose on glucagon release. We also identified glycine, acting via α-cell glycine receptors, as the predominant amino acid stimulator of glucagon release. The results suggest that glycine and GHB provide a counterbalancing receptor-based mechanism for controlling α-cell secretory responses to metabolic fuels.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagon/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Glicina/metabolismo , Células Secretoras de Insulina/metabolismo , Oxibato de Sódio/metabolismo , Adulto , Diabetes Mellitus Tipo 2/patologia , Feminino , Células Secretoras de Glucagon/patologia , Humanos , Células Secretoras de Insulina/patologia , Masculino , Pessoa de Meia-Idade , Receptores de GABA/metabolismo , Receptores de Glicina/metabolismo , Ácido gama-Aminobutírico/metabolismo
8.
Biochem J ; 444(3): 537-51, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22448977

RESUMO

GKAs (glucokinase activators) are promising agents for the therapy of Type 2 diabetes, but little is known about their effects on hepatic intermediary metabolism. We monitored the fate of (13)C-labelled glucose in both a liver perfusion system and isolated hepatocytes. MS and NMR spectroscopy were deployed to measure isotopic enrichment. The results demonstrate that the stimulation of glycolysis by GKA led to numerous changes in hepatic metabolism: (i) augmented flux through the TCA (tricarboxylic acid) cycle, as evidenced by greater incorporation of (13)C into the cycle (anaplerosis) and increased generation of (13)C isotopomers of citrate, glutamate and aspartate (cataplerosis); (ii) lowering of hepatic [Pi] and elevated [ATP], denoting greater phosphorylation potential and energy state; (iii) stimulation of glycogen synthesis from glucose, but inhibition of glycogen synthesis from 3-carbon precursors; (iv) increased synthesis of N-acetylglutamate and consequently augmented ureagenesis; (v) increased synthesis of glutamine, alanine, serine and glycine; and (vi) increased production and outflow of lactate. The present study provides a deeper insight into the hepatic actions of GKAs and uncovers the potential benefits and risks of GKA for treatment of diabetes. GKA improved hepatic bioenergetics, ureagenesis and glycogenesis, but decreased gluconeogenesis with a potential risk of lactic acidosis and fatty liver.


Assuntos
Benzenoacetamidas/farmacologia , Glucoquinase/metabolismo , Hepatócitos/enzimologia , Metabolômica/métodos , Animais , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley
9.
J Biol Chem ; 286(25): 22055-68, 2011 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-21540182

RESUMO

We previously reported that isobutylmethylxanthine (IBMX), a derivative of oxypurine, inhibits citrulline synthesis by an as yet unknown mechanism. Here, we demonstrate that IBMX and other oxypurines containing a 2,6-dione group interfere with the binding of glutamate to the active site of N-acetylglutamate synthetase (NAGS), thereby decreasing synthesis of N-acetylglutamate, the obligatory activator of carbamoyl phosphate synthase-1 (CPS1). The result is reduction of citrulline and urea synthesis. Experiments were performed with (15)N-labeled substrates, purified hepatic CPS1, and recombinant mouse NAGS as well as isolated mitochondria. We also used isolated hepatocytes to examine the action of various oxypurines on ureagenesis and to assess the ameliorating affect of N-carbamylglutamate and/or l-arginine on NAGS inhibition. Among various oxypurines tested, only IBMX, xanthine, or uric acid significantly increased the apparent K(m) for glutamate and decreased velocity of NAGS, with little effect on CPS1. The inhibition of NAGS is time- and dose-dependent and leads to decreased formation of the CPS1-N-acetylglutamate complex and consequent inhibition of citrulline and urea synthesis. However, such inhibition was reversed by supplementation with N-carbamylglutamate. The data demonstrate that xanthine and uric acid, both physiologically occurring oxypurines, inhibit the hepatic synthesis of N-acetylglutamate. An important and novel concept emerging from this study is that xanthine and/or uric acid may have a role in the regulation of ureagenesis and, thus, nitrogen homeostasis in normal and disease states.


Assuntos
Aminoácido N-Acetiltransferase/antagonistas & inibidores , Regulação para Baixo/efeitos dos fármacos , Fígado/metabolismo , Ureia/metabolismo , Ácido Úrico/farmacologia , Xantina/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Aminoácido N-Acetiltransferase/isolamento & purificação , Aminoácido N-Acetiltransferase/metabolismo , Animais , Carbamoil-Fosfato Sintase (Amônia)/isolamento & purificação , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , Citrulina/biossíntese , Relação Dose-Resposta a Droga , Glutamatos/biossíntese , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Cinética , Fígado/citologia , Fígado/enzimologia , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ratos , Ratos Sprague-Dawley
10.
J Vis Exp ; (48)2011 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-21403629

RESUMO

Stable isotopic profiling has long permitted sensitive investigations of the metabolic consequences of genetic mutations and/or pharmacologic therapies in cellular and mammalian models. Here, we describe detailed methods to perform stable isotopic profiling of intermediary metabolism and metabolic flux in the nematode, Caenorhabditis elegans. Methods are described for profiling whole worm free amino acids, labeled carbon dioxide, labeled organic acids, and labeled amino acids in animals exposed to stable isotopes either from early development on nematode growth media agar plates or beginning as young adults while exposed to various pharmacologic treatments in liquid culture. Free amino acids are quantified by high performance liquid chromatography (HPLC) in whole worm aliquots extracted in 4% perchloric acid. Universally labeled (13)C-glucose or 1,6-(13)C(2)-glucose is utilized as the stable isotopic precursor whose labeled carbon is traced by mass spectrometry in carbon dioxide (both atmospheric and dissolved) as well as in metabolites indicative of flux through glycolysis, pyruvate metabolism, and the tricarboxylic acid cycle. Representative results are included to demonstrate effects of isotope exposure time, various bacterial clearing protocols, and alternative worm disruption methods in wild-type nematodes, as well as the relative extent of isotopic incorporation in mitochondrial complex III mutant worms (isp-1(qm150)) relative to wild-type worms. Application of stable isotopic profiling in living nematodes provides a novel capacity to investigate at the whole animal level real-time metabolic alterations that are caused by individual genetic disorders and/or pharmacologic therapies.


Assuntos
Caenorhabditis elegans/metabolismo , Isótopos de Carbono/metabolismo , Glucose/metabolismo , Marcação por Isótopo/métodos , Animais , Caenorhabditis elegans/crescimento & desenvolvimento
11.
Mol Genet Metab ; 100 Suppl 1: S37-41, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20338795

RESUMO

Stable isotopes have been an invaluable adjunct to biomedical research for more than 70years. Indeed, the isotopic approach has revolutionized our understanding of metabolism, revealing it to be an intensely dynamic process characterized by an unending cycle of synthesis and degradation. Isotopic studies have taught us that the urea cycle is intrinsic to such dynamism, since it affords a capacious mechanism by which to eliminate waste nitrogen when rates of protein degradation (or dietary protein intake) are especially high. Isotopes have enabled an appreciation of the degree to which ureagenesis is compromised in patients with urea cycle defects. Indeed, isotopic studies of urea cycle flux correlate well with the severity of cognitive impairment in these patients. Finally, the use of isotopes affords an ideal tool with which to gauge the efficacy of therapeutic interventions to augment residual flux through the cycle.


Assuntos
Marcação por Isótopo/métodos , Ureia/metabolismo , Aminoácido N-Acetiltransferase/deficiência , Aminoácido N-Acetiltransferase/metabolismo , Cloreto de Amônio/administração & dosagem , Cloreto de Amônio/farmacologia , Dióxido de Carbono/metabolismo , Isótopos de Carbono/metabolismo , Humanos , Ureia/sangue
12.
Mol Genet Metab ; 98(4): 325-30, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19660971

RESUMO

We studied the effect on ureagenesis of a single dose of N-carbamylglutamate (NCG) in healthy young adults who received a constant infusion (300 min) of NaH(13)CO(3). Isotope ratio-mass spectrometry was used to measure the appearance of label in [(13)C]urea. At 90 min after initiating the H(13)CO3-infusion each subject took a single dose of NCG (50 mg/kg). In 5/6 studies the administration of NCG increased the formation of [(13)C]urea. Treatment with NCG significantly diminished the concentration of blood alanine, but not that of glutamine or arginine. The blood glucose concentration was unaffected by NCG administration. No untoward side effects were observed. The data indicate that treatment with NCG stimulates ureagenesis and could be useful in clinical settings of acute hyperammonemia of various etiologies.


Assuntos
Glutamatos/administração & dosagem , Glutamatos/farmacologia , Ureia/metabolismo , Adulto , Aminoácidos/sangue , Glicemia/efeitos dos fármacos , Dióxido de Carbono/metabolismo , Isótopos de Carbono , Demografia , Relação Dose-Resposta a Droga , Expiração/efeitos dos fármacos , Feminino , Saúde , Humanos , Infusões Intravenosas , Masculino , Bicarbonato de Sódio/administração & dosagem , Bicarbonato de Sódio/farmacologia , Ureia/sangue , Adulto Jovem
13.
Epilepsia ; 49 Suppl 8: 73-5, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19049594

RESUMO

We hypothesize that one mechanism of the anti-epileptic effect of the ketogenic diet is to alter brain handling of glutamate. According to this formulation, in ketotic brain astrocyte metabolism is more active, resulting in enhanced conversion of glutamate to glutamine. This allows for: (a) more efficient removal of glutamate, the most important excitatory neurotransmitter; and (b) more efficient conversion of glutamine to GABA, the major inhibitory neurotransmitter.


Assuntos
Encéfalo/metabolismo , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Cetose/metabolismo , Ácido gama-Aminobutírico/metabolismo , Animais , Dieta Cetogênica , Humanos
14.
Proc Natl Acad Sci U S A ; 105(48): 18782-7, 2008 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-19033189

RESUMO

Mammalian cells fuel their growth and proliferation through the catabolism of two main substrates: glucose and glutamine. Most of the remaining metabolites taken up by proliferating cells are not catabolized, but instead are used as building blocks during anabolic macromolecular synthesis. Investigations of phosphoinositol 3-kinase (PI3K) and its downstream effector AKT have confirmed that these oncogenes play a direct role in stimulating glucose uptake and metabolism, rendering the transformed cell addicted to glucose for the maintenance of survival. In contrast, less is known about the regulation of glutamine uptake and metabolism. Here, we report that the transcriptional regulatory properties of the oncogene Myc coordinate the expression of genes necessary for cells to engage in glutamine catabolism that exceeds the cellular requirement for protein and nucleotide biosynthesis. A consequence of this Myc-dependent glutaminolysis is the reprogramming of mitochondrial metabolism to depend on glutamine catabolism to sustain cellular viability and TCA cycle anapleurosis. The ability of Myc-expressing cells to engage in glutaminolysis does not depend on concomitant activation of PI3K or AKT. The stimulation of mitochondrial glutamine metabolism resulted in reduced glucose carbon entering the TCA cycle and a decreased contribution of glucose to the mitochondrial-dependent synthesis of phospholipids. These data suggest that oncogenic levels of Myc induce a transcriptional program that promotes glutaminolysis and triggers cellular addiction to glutamine as a bioenergetic substrate.


Assuntos
Metabolismo Energético/fisiologia , Glutamina/metabolismo , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Glucose/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais/fisiologia
15.
J Biol Chem ; 283(25): 17238-49, 2008 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-18445600

RESUMO

Pancreatic beta cells are hyper-responsive to amino acids but have decreased glucose sensitivity after deletion of the sulfonylurea receptor 1 (SUR1) both in man and mouse. It was hypothesized that these defects are the consequence of impaired integration of amino acid, glucose, and energy metabolism in beta cells. We used gas chromatography-mass spectrometry methodology to study intermediary metabolism of SUR1 knock-out (SUR1(-/-)) and control mouse islets with d-[U-(13)C]glucose as substrate and related the results to insulin secretion. The levels and isotope labeling of alanine, aspartate, glutamate, glutamine, and gamma-aminobutyric acid (GABA) served as indicators of intermediary metabolism. We found that the GABA shunt of SUR1(-/-) islets is blocked by about 75% and showed that this defect is due to decreased glutamate decarboxylase synthesis, probably caused by elevated free intracellular calcium. Glutaminolysis stimulated by the leucine analogue d,l-beta-2-amino-2-norbornane-carboxylic acid was, however, enhanced in SUR1(-/-) and glyburide-treated SUR1(+/+) islets. Glucose oxidation and pyruvate cycling was increased in SUR1(-/-) islets at low glucose but was the same as in controls at high glucose. Malic enzyme isoforms 1, 2, and 3, involved in pyruvate cycling, were all expressed in islets. High glucose lowered aspartate and stimulated glutamine synthesis similarly in controls and SUR1(-/-) islets. The data suggest that the interruption of the GABA shunt and the lack of glucose regulation of pyruvate cycling may cause the glucose insensitivity of the SUR1(-/-) islets but that enhanced basal pyruvate cycling, lowered GABA shunt flux, and enhanced glutaminolytic capacity may sensitize the beta cells to amino acid stimulation.


Assuntos
Trifosfato de Adenosina/química , Glucose/metabolismo , Glutamina/química , Potássio/química , Piruvatos/química , Ácido gama-Aminobutírico/metabolismo , Aminoácidos/química , Animais , Cromatografia Gasosa-Espectrometria de Massas/métodos , Genótipo , Glutamato Descarboxilase/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Oxigênio/metabolismo
16.
J Biol Chem ; 283(22): 15063-71, 2008 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-18375388

RESUMO

We previously showed that agmatine stimulated hepatic ureagenesis. In this study, we sought to determine whether the action of agmatine is mediated via cAMP signaling. A pilot experiment demonstrated that the phosphodiesterase inhibitor, 3-isobutylmethylxanthine (IBMX), inhibited urea synthesis albeit increased [cAMP]. Thus, we hypothesized that IBMX inhibits hepatic urea synthesis independent of [cAMP]. We further theorized that agmatine would negate the IBMX action and improve ureagenesis. Experiments were carried out with isolated mitochondria and (15)NH(4)Cl to trace [(15)N]citrulline production or [5-(15)N]glutamine and a rat liver perfusion system to trace ureagenesis. The results demonstrate that IBMX induced the following: (i) inhibition of the mitochondrial respiratory chain and diminished O(2) consumption during liver perfusion; (ii) depletion of the phosphorylation potential and overall hepatic energetic capacity; (iii) inhibition of [(15)N]citrulline synthesis; and (iv) inhibition of urea output in liver perfusion with little effect on [N-acetylglutamate]. The results indicate that IBMX directly and specifically inhibited complex I of the respiratory chain and carbamoyl-phosphate synthase-I (CPS-I), with an EC(50) about 0.6 mm despite a significant elevation of hepatic [cAMP]. Perfusion of agmatine with IBMX stimulated O(2) consumption, restored hepatic phosphorylation potential, and significantly stimulated ureagenesis. The action of agmatine may signify a cascade effect initiated by increased oxidative phosphorylation and greater ATP synthesis. In addition, agmatine may prevent IBMX from binding to one or more active site(s) of CPS-I and thus protect against inhibition of CPS-I. Together, the data may suggest a new experimental application of IBMX in studies of CPS-I malfunction and the use of agmatine as intervention therapy.


Assuntos
1-Metil-3-Isobutilxantina/farmacologia , Agmatina/farmacologia , Fígado/metabolismo , Mitocôndrias Hepáticas/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Ureia/metabolismo , Animais , Carbamoil-Fosfato Sintase (Amônia)/metabolismo , AMP Cíclico/metabolismo , Transporte de Elétrons/efeitos dos fármacos , Masculino , Consumo de Oxigênio/efeitos dos fármacos , Perfusão , Diester Fosfórico Hidrolases/metabolismo , Ratos , Ratos Sprague-Dawley
17.
Pediatr Res ; 64(2): 213-7, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18414145

RESUMO

N-acetylglutamate (NAG) is an endogenous essential cofactor for conversion of ammonia to urea in the liver. Deficiency of NAG causes hyperammonemia and occurs because of inherited deficiency of its producing enzyme, NAG synthase (NAGS), or interference with its function by short fatty acid derivatives. N-carbamylglutamate (NCG) can ameliorate hyperammonemia from NAGS deficiency and propionic and methylmalonic acidemia. We developed a stable isotope (13)C tracer method to measure ureagenesis and to evaluate the effect of NCG in humans. Seventeen healthy adults were investigated for the incorporation of (13)C label into urea. [(13)C]urea appeared in the blood within minutes, reaching maximum by 100 min, whereas breath (13)CO(2) reached a maximum by 60 min. A patient with NAGS deficiency showed very little urea labeling before treatment with NCG and normal labeling thereafter. Correspondingly, plasma levels of ammonia and glutamine decreased markedly and urea tripled after NCG treatment. Similarly, in a patient with propionic acidemia, NCG treatment resulted in a marked increase in urea labeling and decrease in glutamine, alanine, and glycine. These results provide a reliable method for measuring the effect of NCG on nitrogen metabolism and strongly suggest that NCG could be an effective treatment for inherited and secondary NAGS deficiency.


Assuntos
Glutamatos/deficiência , Glutamatos/farmacologia , Doenças Metabólicas/sangue , Propionatos/sangue , Ureia/sangue , Acetilcoenzima A/metabolismo , Adulto , Aminoácidos/sangue , Amônia/sangue , Biomarcadores/sangue , Dióxido de Carbono/metabolismo , Isótopos de Carbono , Criança , Estudos de Viabilidade , Feminino , Glutamatos/metabolismo , Glutamatos/uso terapêutico , Humanos , Masculino , Doenças Metabólicas/tratamento farmacológico , Pessoa de Meia-Idade
18.
Proc Natl Acad Sci U S A ; 104(49): 19345-50, 2007 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-18032601

RESUMO

Tumor cell proliferation requires rapid synthesis of macromolecules including lipids, proteins, and nucleotides. Many tumor cells exhibit rapid glucose consumption, with most of the glucose-derived carbon being secreted as lactate despite abundant oxygen availability (the Warburg effect). Here, we used 13C NMR spectroscopy to examine the metabolism of glioblastoma cells exhibiting aerobic glycolysis. In these cells, the tricarboxylic acid (TCA) cycle was active but was characterized by an efflux of substrates for use in biosynthetic pathways, particularly fatty acid synthesis. The success of this synthetic activity depends on activation of pathways to generate reductive power (NADPH) and to restore oxaloacetate for continued TCA cycle function (anaplerosis). Surprisingly, both these needs were met by a high rate of glutamine metabolism. First, conversion of glutamine to lactate (glutaminolysis) was rapid enough to produce sufficient NADPH to support fatty acid synthesis. Second, despite substantial mitochondrial pyruvate metabolism, pyruvate carboxylation was suppressed, and anaplerotic oxaloacetate was derived from glutamine. Glutamine catabolism was accompanied by secretion of alanine and ammonia, such that most of the amino groups from glutamine were lost from the cell rather than incorporated into other molecules. These data demonstrate that transformed cells exhibit a high rate of glutamine consumption that cannot be explained by the nitrogen demand imposed by nucleotide synthesis or maintenance of nonessential amino acid pools. Rather, glutamine metabolism provides a carbon source that facilitates the cell's ability to use glucose-derived carbon and TCA cycle intermediates as biosynthetic precursors.


Assuntos
Neoplasias Encefálicas/metabolismo , Transformação Celular Neoplásica/metabolismo , Glioblastoma/metabolismo , Glutamina/metabolismo , Glicólise , Aerobiose , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/patologia , Ciclo do Ácido Cítrico , Glioblastoma/patologia , Glucose/metabolismo , Humanos , Nucleotídeos/biossíntese , Biossíntese de Proteínas
19.
Annu Rev Nutr ; 27: 415-30, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17444813

RESUMO

In many epileptic patients, anticonvulsant drugs either fail adequately to control seizures or they cause serious side effects. An important adjunct to pharmacologic therapy is the ketogenic diet, which often improves seizure control, even in patients who respond poorly to medications. The mechanisms that explain the therapeutic effect are incompletely understood. Evidence points to an effect on brain handling of amino acids, especially glutamic acid, the major excitatory neurotransmitter of the central nervous system. The diet may limit the availability of oxaloacetate to the aspartate aminotransferase reaction, an important route of brain glutamate handling. As a result, more glutamate becomes accessible to the glutamate decarboxylase reaction to yield gamma-aminobutyric acid (GABA), the major inhibitory neurotransmitter and an important antiseizure agent. In addition, the ketogenic diet appears to favor the synthesis of glutamine, an essential precursor to GABA. This occurs both because ketone body carbon is metabolized to glutamine and because in ketosis there is increased consumption of acetate, which astrocytes in the brain quickly convert to glutamine. The ketogenic diet also may facilitate mechanisms by which the brain exports to blood compounds such as glutamine and alanine, in the process favoring the removal of glutamate carbon and nitrogen.


Assuntos
Aminoácidos/metabolismo , Encéfalo/metabolismo , Corpos Cetônicos/metabolismo , Cetose/metabolismo , Convulsões/metabolismo , Anticonvulsivantes/uso terapêutico , Terapia Combinada , Dieta , Ácido Glutâmico/metabolismo , Humanos , Convulsões/dietoterapia , Convulsões/tratamento farmacológico , Resultado do Tratamento
20.
Cancer Res ; 66(15): 7824-31, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16885387

RESUMO

The efficacy of ifosfamide (IFO), an antineoplastic drug, is severely limited by a high incidence of nephrotoxicity of unknown etiology. We hypothesized that inhibition of complex I (C-I) by chloroacetaldehyde (CAA), a metabolite of IFO, is the chief cause of nephrotoxicity, and that agmatine (AGM), which we found to augment mitochondrial oxidative phosphorylation and beta-oxidation, would prevent nephrotoxicity. Our model system was isolated mitochondria obtained from the kidney cortex of rats treated with IFO or IFO + AGM. Oxidative phosphorylation was determined with electron donors specific to complexes I, II, III, or IV (C-I, C-II, C-III, or C-IV, respectively). A parallel study was done with (13)C-labeled pyruvate to assess metabolic dysfunction. Ifosfamide treatment significantly inhibited oxidative phosphorylation with only C-I substrates. Inhibition of C-I was associated with a significant elevation of [NADH], depletion of [NAD], and decreased flux through pyruvate dehydrogenase and the TCA cycle. However, administration of AGM with IFO increased [cyclic AMP (cAMP)] and prevented IFO-induced inhibition of C-I. In vitro studies with various metabolites of IFO showed that only CAA inhibited C-I, even with supplementation with 2-mercaptoethane sulfonic acid. Following IFO treatment daily for 5 days with 50 mg/kg, the level of CAA in the renal cortex was approximately 15 micromol/L. Taken together, these observations support the hypothesis that CAA is accumulated in renal cortex and is responsible for nephrotoxicity. AGM may be protective by increasing tissue [cAMP], which phosphorylates NADH:oxidoreductase. The current findings may have an important implication for the prevention of IFO-induced nephrotoxicity and/or mitochondrial diseases secondary to defective C-I.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Ifosfamida/toxicidade , Nefropatias/induzido quimicamente , Nefropatias/prevenção & controle , Acetaldeído/análogos & derivados , Acetaldeído/farmacocinética , Agmatina/farmacologia , Animais , Interações Medicamentosas , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Complexo I de Transporte de Elétrons/metabolismo , Ifosfamida/farmacocinética , Córtex Renal/enzimologia , Córtex Renal/metabolismo , Nefropatias/enzimologia , Masculino , Fosforilação Oxidativa/efeitos dos fármacos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA