Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Nature ; 615(7952): 490-498, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36890227

RESUMO

Metabolic rewiring underlies the effector functions of macrophages1-3, but the mechanisms involved remain incompletely defined. Here, using unbiased metabolomics and stable isotope-assisted tracing, we show that an inflammatory aspartate-argininosuccinate shunt is induced following lipopolysaccharide stimulation. The shunt, supported by increased argininosuccinate synthase (ASS1) expression, also leads to increased cytosolic fumarate levels and fumarate-mediated protein succination. Pharmacological inhibition and genetic ablation of the tricarboxylic acid cycle enzyme fumarate hydratase (FH) further increases intracellular fumarate levels. Mitochondrial respiration is also suppressed and mitochondrial membrane potential increased. RNA sequencing and proteomics analyses demonstrate that there are strong inflammatory effects resulting from FH inhibition. Notably, acute FH inhibition suppresses interleukin-10 expression, which leads to increased tumour necrosis factor secretion, an effect recapitulated by fumarate esters. Moreover, FH inhibition, but not fumarate esters, increases interferon-ß production through mechanisms that are driven by mitochondrial RNA (mtRNA) release and activation of the RNA sensors TLR7, RIG-I and MDA5. This effect is recapitulated endogenously when FH is suppressed following prolonged lipopolysaccharide stimulation. Furthermore, cells from patients with systemic lupus erythematosus also exhibit FH suppression, which indicates a potential pathogenic role for this process in human disease. We therefore identify a protective role for FH in maintaining appropriate macrophage cytokine and interferon responses.


Assuntos
Fumarato Hidratase , Interferon beta , Macrófagos , Mitocôndrias , RNA Mitocondrial , Humanos , Argininossuccinato Sintase/metabolismo , Ácido Argininossuccínico/metabolismo , Ácido Aspártico/metabolismo , Respiração Celular , Citosol/metabolismo , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/genética , Fumarato Hidratase/metabolismo , Fumaratos/metabolismo , Interferon beta/biossíntese , Interferon beta/imunologia , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Lúpus Eritematoso Sistêmico/enzimologia , Macrófagos/enzimologia , Macrófagos/imunologia , Macrófagos/metabolismo , Potencial da Membrana Mitocondrial , Metabolômica , Mitocôndrias/genética , Mitocôndrias/metabolismo , RNA Mitocondrial/metabolismo
2.
Mol Cell ; 51(2): 236-48, 2013 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-23747014

RESUMO

The tricarboxylic acid cycle enzyme fumarate hydratase (FH) has been identified as a tumor suppressor in a subset of human renal cell carcinomas. Human FH-deficient cancer cells display high fumarate concentration and ROS levels along with activation of HIF-1. The underlying mechanisms by which FH loss increases ROS and HIF-1 are not fully understood. Here, we report that glutamine-dependent oxidative citric acid cycle metabolism is required to generate fumarate and increase ROS and HIF-1 levels. Accumulated fumarate directly bonds the antioxidant glutathione in vitro and in vivo to produce the metabolite succinated glutathione (GSF). GSF acts as an alternative substrate to glutathione reductase to decrease NADPH levels and enhance mitochondrial ROS and HIF-1 activation. Increased ROS also correlates with hypermethylation of histones in these cells. Thus, fumarate serves as a proto-oncometabolite by binding to glutathione which results in the accumulation of ROS.


Assuntos
Carcinoma de Células Renais/metabolismo , Fumaratos/metabolismo , Glutationa/metabolismo , Neoplasias Renais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Carcinoma de Células Renais/patologia , Cromatografia Líquida , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/genética , Fumarato Hidratase/metabolismo , Glutationa Redutase/metabolismo , Histona Desmetilases/metabolismo , Histonas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Immunoblotting , Neoplasias Renais/patologia , NADP/metabolismo , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Consumo de Oxigênio , RNA Interferente Pequeno/genética , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Células Tumorais Cultivadas
3.
Artigo em Inglês | MEDLINE | ID: mdl-31332064

RESUMO

In the last decade, carbon monoxide-releasing molecules (CORMs) have been shown to act against several pathogens and to be promising antimicrobials. However, the understanding of the mode of action and reactivity of these compounds on bacterial cells is still deficient. In this work, we used a metabolomics approach to probe the toxicity of the ruthenium(II) complex Ru(CO)3Cl(glycinate) (CORM-3) on Escherichia coli By resorting to 1H nuclear magnetic resonance, mass spectrometry, and enzymatic activities, we show that CORM-3-treated E. coli accumulates larger amounts of glycolytic intermediates, independently of the oxygen growth conditions. The work provides several evidences that CORM-3 inhibits glutamate synthesis and the iron-sulfur enzymes of the tricarboxylic acid (TCA) cycle and that the glycolysis pathway is triggered in order to establish an energy and redox homeostasis balance. Accordingly, supplementation of the growth medium with fumarate, α-ketoglutarate, glutamate, and amino acids cancels the toxicity of CORM-3. Importantly, inhibition of the iron-sulfur enzymes glutamate synthase, aconitase, and fumarase is only observed for compounds that liberate carbon monoxide. Altogether, this work reveals that the antimicrobial action of CORM-3 results from intracellular glutamate deficiency and inhibition of nitrogen and TCA cycles.


Assuntos
Antibacterianos/farmacologia , Monóxido de Carbono/farmacologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Regulação Bacteriana da Expressão Gênica , Nitrogênio/metabolismo , Compostos Organometálicos/farmacologia , Aconitato Hidratase/antagonistas & inibidores , Aconitato Hidratase/genética , Aconitato Hidratase/metabolismo , Antibacterianos/química , Monóxido de Carbono/química , Ciclo do Ácido Cítrico/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/genética , Fumarato Hidratase/metabolismo , Fumaratos/metabolismo , Glutamato Sintase/antagonistas & inibidores , Glutamato Sintase/genética , Glutamato Sintase/metabolismo , Ácido Glutâmico/metabolismo , Glicólise/efeitos dos fármacos , Glicólise/genética , Ácidos Cetoglutáricos/metabolismo , Espectroscopia de Ressonância Magnética , Metabolômica/métodos , Compostos Organometálicos/química , Oxirredução
4.
Proc Natl Acad Sci U S A ; 113(27): 7503-8, 2016 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-27325754

RESUMO

Enzymes in essential metabolic pathways are attractive targets for the treatment of bacterial diseases, but in many cases, the presence of homologous human enzymes makes them impractical candidates for drug development. Fumarate hydratase, an essential enzyme in the tricarboxylic acid (TCA) cycle, has been identified as one such potential therapeutic target in tuberculosis. We report the discovery of the first small molecule inhibitor, to our knowledge, of the Mycobacterium tuberculosis fumarate hydratase. A crystal structure at 2.0-Å resolution of the compound in complex with the protein establishes the existence of a previously unidentified allosteric regulatory site. This allosteric site allows for selective inhibition with respect to the homologous human enzyme. We observe a unique binding mode in which two inhibitor molecules interact within the allosteric site, driving significant conformational changes that preclude simultaneous substrate and inhibitor binding. Our results demonstrate the selective inhibition of a highly conserved metabolic enzyme that contains identical active site residues in both the host and the pathogen.


Assuntos
Fumarato Hidratase/metabolismo , Mycobacterium tuberculosis/enzimologia , Regulação Alostérica , Cristalografia por Raios X , Fluorescência , Fumarato Hidratase/antagonistas & inibidores
5.
Malar J ; 16(1): 247, 2017 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-28606087

RESUMO

BACKGROUND: Aspartate, which is converted from oxaloacetate (OAA) by aspartate aminotransferase, is considered an important precursor for purine salvage and pyrimidine de novo biosynthesis, and is thus indispensable for the growth of Plasmodium parasites at the asexual blood stages. OAA can be produced in malaria parasites via two routes: (i) from phosphoenolpyruvate (PEP) by phosphoenolpyruvate carboxylase (PEPC) in the cytosol, or (ii) from fumarate by consecutive reactions catalyzed by fumarate hydratase (FH) and malate:quinone oxidoreductase (MQO) in the mitochondria of malaria parasites. Although PEPC-deficient Plasmodium falciparum and Plasmodium berghei (rodent malaria) parasites show a growth defect, the mutant P. berghei can still cause experimental cerebral malaria (ECM) with similar dynamics to wild-type parasites. In contrast, the importance of FH and MQO for parasite viability, growth and virulence is not fully understood because no FH- and MQO-deficient P. falciparum has been established. In this study, the role of FH and MQO in the pathogenicity of asexual-blood-stage Plasmodium parasites causing cerebral malaria was examined. RESULTS: First, FH- and MQO-deficient parasites were generated by inserting a luciferase-expressing cassette into the fh and mqo loci in the genome of P. berghei ANKA strain. Second, the viability of FH-deficient and MQO-deficient parasites that express luciferase was determined by measuring luciferase activity, and the effect of FH or MQO deficiency on the development of ECM was examined. While the viability of FH-deficient P. berghei was comparable to that of control parasites, MQO-deficient parasites exhibited considerably reduced viability. FH activity derived from erythrocytes was also detected. This result and the absence of phenotype in FH-deficient P. berghei parasites suggest that fumarate can be metabolized to malate by host or parasite FH in P. berghei-infected erythrocytes. Furthermore, although the growth of FH- and MQO-deficient parasites was impaired, the development of ECM was suppressed only in mice infected with MQO-deficient parasites. CONCLUSIONS: These findings suggest that MQO-mediated mitochondrial functions are required for development of ECM of asexual-blood-stage Plasmodium parasites.


Assuntos
Malária Cerebral/prevenção & controle , Mitocôndrias/enzimologia , Oxirredutases/antagonistas & inibidores , Plasmodium berghei/enzimologia , Animais , Barreira Hematoencefálica/metabolismo , Eritrócitos/parasitologia , Feminino , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/deficiência , Fumarato Hidratase/fisiologia , Fumaratos/metabolismo , Malatos/metabolismo , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/fisiologia , Ácido Oxaloacético/metabolismo , Oxirredutases/deficiência , Oxirredutases/fisiologia , Plasmodium berghei/genética , Plasmodium berghei/crescimento & desenvolvimento , Organismos Livres de Patógenos Específicos
6.
J Am Chem Soc ; 137(2): 564-7, 2015 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-25469852

RESUMO

Development of cell-permeable small molecules that target enzymes involved in energy metabolism remains important yet challenging. We describe here the discovery of a new class of compounds with a nutrient-dependent cytotoxicity profile that arises from pharmacological inhibition of fumarate hydratase (also known as fumarase). This finding was enabled by a high-throughput screen of a diverse chemical library in a panel of human cancer cell lines cultured under different growth conditions, followed by subsequent structure-activity optimization and target identification. While the highest cytotoxicity was observed under low glucose concentrations, the antiproliferative activities and inhibition of oxygen consumption rates in cells were distinctly different from those displayed by typical inhibitors of mitochondrial oxidative phosphorylation. The use of a photoaffinity labeling strategy identified fumarate hydratase as the principal pharmacological target. Final biochemical studies confirmed dose-dependent, competitive inhibition of this enzyme in vitro, which was fully consistent with the initially observed growth inhibitory activity. Our work demonstrates how the phenotypic observations combined with a successful target identification strategy can yield a useful class of pharmacological inhibitors of an enzyme involved in the operation of tricarboxylic acid cycle.


Assuntos
Inibidores Enzimáticos/farmacologia , Fumarato Hidratase/antagonistas & inibidores , Linhagem Celular Tumoral , Ciclo do Ácido Cítrico/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Metabolismo Energético/efeitos dos fármacos , Inibidores Enzimáticos/toxicidade , Fumarato Hidratase/metabolismo , Glucose/metabolismo , Ensaios de Triagem em Larga Escala , Humanos
7.
Cancer Cell ; 8(2): 143-53, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16098467

RESUMO

Individuals with hemizygous germline fumarate hydratase (FH) mutations are predisposed to renal cancer. These tumors predominantly exhibit functional inactivation of the remaining wild-type allele, implicating FH inactivation as a tumor-promoting event. Hypoxia-inducible factors are expressed in many cancers and are increased in clear cell renal carcinomas. Under normoxia, the HIFs are labile due to VHL-dependent proteasomal degradation, but stabilization occurs under hypoxia due to inactivation of HIF prolyl hydroxylase (HPH), which prevents HIF hydroxylation and VHL recognition. We demonstrate that FH inhibition, together with elevated intracellular fumarate, coincides with HIF upregulation. Further, we show that fumarate acts as a competitive inhibitor of HPH. These data delineate a novel fumarate-dependent pathway for regulating HPH activity and HIF protein levels.


Assuntos
Carcinoma de Células Renais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fumarato Hidratase/genética , Fumaratos/metabolismo , Neoplasias Renais/metabolismo , Leiomiomatose/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Alelos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/genética , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Feminino , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/metabolismo , Fumaratos/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Ácidos Cetoglutáricos/farmacologia , Neoplasias Renais/enzimologia , Neoplasias Renais/genética , Leiomiomatose/enzimologia , Leiomiomatose/genética , Masculino , Pessoa de Meia-Idade , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Síndrome , Fatores de Transcrição/análise , Fatores de Transcrição/genética , Regulação para Cima
8.
Biometals ; 25(1): 95-102, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21818585

RESUMO

Helicobacter pylori causes various gastric diseases, such as gastritis, peptic ulcerations and gastric cancer. Triple therapy combining bismuth compounds with two antibiotics is the cornerstone of the treatment of H. pylori infections. Up to now, the molecular mechanisms by which bismuth inhibits the growth of H. pylori are far from clear. In the bacterial tricarboxylic acid (TCA) cycle, fumarase catalyses the reversible hydration of fumarate to malic acid. Our previous proteomic work indicated that fumarase was capable of bismuth-binding. The interactions as well as the inhibitory effects of bismuth to fumarase have been characterized in this study. The titration of bismuth showed that each fumarase monomer binds one mol equiv of Bi(3+), with negligible secondary structural change. Bismuth-binding results in a near stoichiometric inactivation of the enzyme, leading to an apparent non-competitive mechanism as reflected by the Lineweaver-Burk plots. Our collective data indicate that the TCA cycle is a potential molecular target of bismuth drugs in H. pylori.


Assuntos
Proteínas de Bactérias/metabolismo , Bismuto/farmacologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Fumarato Hidratase/antagonistas & inibidores , Helicobacter pylori/efeitos dos fármacos , Helicobacter pylori/enzimologia , Proteínas de Bactérias/genética , Bismuto/uso terapêutico , Fumarato Hidratase/química , Fumarato Hidratase/genética , Infecções por Helicobacter/tratamento farmacológico , Humanos , Modelos Moleculares , Estrutura Terciária de Proteína
9.
J Med Chem ; 62(23): 10586-10604, 2019 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-31517489

RESUMO

With the growing worldwide prevalence of antibiotic-resistant strains of tuberculosis (TB), new targets are urgently required for the development of treatments with novel modes of action. Fumarate hydratase (fumarase), a vulnerable component of the citric acid cycle in Mycobacterium tuberculosis (Mtb), is a metabolic target that could satisfy this unmet demand. A key challenge in the targeting of Mtb fumarase is its similarity to the human homolog, which shares an identical active site. A potential solution to this selectivity problem was previously found in a high-throughput screening hit that binds in a nonconserved allosteric site. In this work, a structure-activity relationship study was carried out with the determination of further structural biology on the lead series, affording derivatives with sub-micromolar inhibition. Further, the screening of this series against Mtb in vitro identified compounds with potent minimum inhibitory concentrations.


Assuntos
Antituberculosos/química , Antituberculosos/farmacologia , Sistemas de Liberação de Medicamentos , Fumarato Hidratase/antagonistas & inibidores , Mycobacterium tuberculosis/efeitos dos fármacos , Sítios de Ligação , Fumarato Hidratase/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Mycobacterium tuberculosis/enzimologia , Conformação Proteica , Relação Estrutura-Atividade
10.
Cell Death Dis ; 10(6): 413, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138787

RESUMO

Gastric cancer (GC) is one of the most common malignancies worldwide. Due to the low rate of early detection, most GC patients were diagnosed as advance stages and had poor response to chemotherapy. Some studies found that Fumarate hydratase (FH) participated in the DNA damage response and its deficiency was associated with tumorigenesis in some cancers. In this study, we investigated the relationship between FH and cisplatin (CDDP) sensitivity in GC cell lines. We found that FH was the most significant gene which induced by CDDP treatment and the suppression of FH could enhance the cytotoxicity of CDDP. Miconazole Nitrate (MN) could inhibit FH activity and enhance the effect of CDDP in vitro and in vivo. We also investigated the significance of expression of FH in GC tissues. The FH expression, which was higher in GC tissues than in noncancerous tissues, was negatively associated with the prognosis of patients. Together, these results revealed that FH is a reliable indicator for response to CDDP treatment in GC and the inhibition of FH may be a potential strategy to improve the effects of CDDP-based chemotherapy.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/uso terapêutico , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Quimioterapia Combinada , Feminino , Fumarato Hidratase/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Prognóstico , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/genética , Transplante Heterólogo
11.
Int J Biol Macromol ; 102: 42-51, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28359888

RESUMO

Nifurtimox and benznidazole represent the only treatments options available targeting Chagas disease, the most important parasitic infection in the Americas. However, use of these is problematic as they are toxic and ineffective against the more severe stages of the disease. In this work, we used a multidisciplinary approach to characterise the fumarases from Trypanosoma cruzi, the causative agent of Chagas Disease. We showed this trypanosome expresses cytosolic and mitochondrial fumarases that via an iron-sulfur cluster mediate the reversible conversion of fumarate to S-malate. Based on sequence, biochemical properties and co-factor binding, both T. cruzi proteins share characteristics with class I fumarases, enzymes found in bacteria and some other protozoa but absent from humans, that possess class II isoforms instead. Gene disruption suggested that although the cytosolic or mitochondrial fumarase activities are individually dispensable their combined activity is essential for parasite viability. Finally, based on the mechanistic differences with the human (host) fumarase, we designed and validated a selective inhibitor targeting the parasite enzyme. This study showed that T. cruzi fumarases should be exploited as targets for the development of new chemotherapeutic interventions against Chagas disease.


Assuntos
Fumarato Hidratase/metabolismo , Trypanosoma cruzi/enzimologia , Citosol/enzimologia , Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/química , Mitocôndrias/enzimologia , Transporte Proteico , Trypanosoma cruzi/citologia
12.
Bioanalysis ; 8(7): 661-75, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26978279

RESUMO

BACKGROUND: Fumaric acid is a commonly used excipient in pharmaceutical products. It is not known if its presence may lead to fluctuation of endogenous fumarate levels. An LC-MS/MS method was developed and validated to quantify fumarate in support of a toxicokinetics study. RESULTS: Stability evaluation showed that endogenous fumarate was stable for 6 h at room temperature, while exogenously added fumaric acid was converted to malate within 1 h due to the presence of fumarase. Citric acid, a fumarase inhibitor, prevented the conversion of added fumaric acid in rat plasma. CONCLUSION: The method was validated in citric acid stabilized rat plasma using a surrogate matrix approach. A discrepancy in stability was observed between endogenous fumarate and exogenously added fumaric acid.


Assuntos
Cromatografia Líquida de Alta Pressão , Fumaratos/sangue , Espectrometria de Massas em Tandem , Animais , Radioisótopos de Carbono/química , Cromatografia Líquida de Alta Pressão/normas , Ácido Cítrico/química , Ácido Cítrico/metabolismo , Estabilidade de Medicamentos , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/metabolismo , Fumaratos/normas , Marcação por Isótopo , Malatos/análise , Malatos/metabolismo , Controle de Qualidade , Ratos , Espectrometria de Massas em Tandem/normas , Temperatura
13.
Cancer Res ; 76(19): 5743-5755, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27302170

RESUMO

Chromatin modification is pivotal to the epithelial-mesenchymal transition (EMT), which confers potent metastatic potential to cancer cells. Here, we report a role for the chromatin remodeling factor lymphoid-specific helicase (LSH) in nasopharyngeal carcinoma (NPC), a prevalent cancer in China. LSH expression was increased in NPC, where it was controlled by the Epstein-Barr virus-encoded protein LMP1. In NPC cells in vitro and in vivo, LSH promoted cancer progression in part by regulating expression of fumarate hydratase (FH), a core component of the tricarboxylic acid cycle. LSH bound to the FH promoter, recruiting the epigenetic silencer factor G9a to repress FH transcription. Clinically, we found that the concentration of TCA intermediates in NPC patient sera was deregulated in the presence of LSH. RNAi-mediated silencing of FH mimicked LSH overexpression, establishing FH as downstream mediator of LSH effects. The TCA intermediates α-KG and citrate potentiated the malignant character of NPC cells, in part by altering IKKα-dependent EMT gene expression. In this manner, LSH furthered malignant progression of NPC by modifying cancer cell metabolism to support EMT. Cancer Res; 76(19); 5743-55. ©2016 AACR.


Assuntos
Carcinoma/etiologia , Montagem e Desmontagem da Cromatina , DNA Helicases/fisiologia , Fumarato Hidratase/antagonistas & inibidores , Neoplasias Nasofaríngeas/etiologia , Animais , Carcinoma/enzimologia , Linhagem Celular Tumoral , Ácido Cítrico/farmacologia , Ciclo do Ácido Cítrico , Progressão da Doença , Transição Epitelial-Mesenquimal , Fumarato Hidratase/metabolismo , Humanos , Ácidos Cetoglutáricos/farmacologia , Camundongos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/enzimologia , Proteína da Zônula de Oclusão-1/análise
14.
Biochim Biophys Acta ; 566(2): 397-9, 1979 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-570424

RESUMO

Active band sedimentation studies of pig heart fumarase indicate that the enzyme is predominantly tetrameric at enzyme concentrations between 0.0125 and 0.25 mg/ml and at a fumarate concentration of 2.5 mM. At enzyme concentrations of 0.25--1.0 mg/ml and fumarate concentrations known to activate and inhibit the enzyme, the sedimentation band of fumarase becomes disperse and indicates the presence of polymers greater than tetramers.


Assuntos
Fumarato Hidratase , Miocárdio/enzimologia , Animais , Ativação Enzimática , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/isolamento & purificação , Fumaratos/farmacologia , Substâncias Macromoleculares , Suínos , Ultracentrifugação
15.
Biochim Biophys Acta ; 1587(1): 31-5, 2002 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-12009421

RESUMO

Ibuprofen and its major metabolites were incubated with catalase and fumarase, in the presence of protein-modifying biomolecules, to explore the mode of action of ibuprofen in protection against cataract. Both 2 and 10 mM ibuprofen/metabolites protected catalase against fructose-, cyanate- and prednisolone-induced inactivation; the carboxy-metabolite gave the highest protection (31%). The 2 mM ibuprofen/metabolites protected fumarase against fructose- and cyanate-induced inactivation by up to 26%, but had no effect on prednisolone-induced inactivation. Ibuprofen/metabolites did not bind to catalase or fumarase. They penetrated into the lens in vitro. When in the lens, the metabolites may reduce the risk of cataract by protecting lenticular enzymes.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Catalase/metabolismo , Fumarato Hidratase/metabolismo , Ibuprofeno/farmacologia , Animais , Biotransformação , Catalase/antagonistas & inibidores , Catarata/prevenção & controle , Bovinos , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Fumarato Hidratase/antagonistas & inibidores , Ibuprofeno/química , Ibuprofeno/metabolismo , Técnicas In Vitro , Cristalino/química , Cristalino/metabolismo , Estrutura Molecular , Permeabilidade
16.
Biochimie ; 57(2): 131-8, 1975.
Artigo em Inglês | MEDLINE | ID: mdl-237577

RESUMO

The catalysis of the hydration of fumarate and deshydration of L - malate by chicken fumarase was measured spectrophotometrically over a range of substrate concentrations from 4 times 10(-3) M to 8 times 10(-5) M for fumarate and from 8 times 10(-2) M to 10(-3) M for L - malate. For the forward and reverse reactions, linear Lineweaver and Burk plots were obtained. The Michaelis constants and the maximum initial velocities for both substrates were determined and the Haldane relation was found to be obeyed. The effect of pH on activity was investigated over a pH range from 5.5 to 9.0 and the data indicate the presence, in the active site, of two ionizable groups, one in the acidic form and one in the basic form. The values of the ionization constants, determined for the enzyme - substrate complexes, agree closely with the ones obtained for the porcine enzyme. The mode of action of twenty-four structural analogs on the initial velocity of the dehydration of L-malate, by chicken fumarase was examined. From these studies, two regions positively charged appear necessary for the effective binding of the carboxylates of the substrates and competitive inhibitors to the active center. Moreover, the data suggest the presence of an additional group, in the catalytic site of chicken fumarase, that stabilizes the carbon-carbon double bond common to fumarate and its structural analogs. Finally, from the comparison of the kinetic properties of the chicken and pig fumarases, it may be concluded that the catalytic mechanism of the homologous enzymes are very similar, if not identical.


Assuntos
Galinhas/metabolismo , Fumarato Hidratase/metabolismo , Hidroliases/metabolismo , Miocárdio/enzimologia , Animais , Sítios de Ligação , Ligação Competitiva , Relação Dose-Resposta a Droga , Fumarato Hidratase/antagonistas & inibidores , Fumaratos/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Malatos/metabolismo , Modelos Biológicos , Ligação Proteica , Relação Estrutura-Atividade , Succinatos/metabolismo , Suínos , Tartaratos/farmacologia , Termodinâmica
17.
J Biochem ; 84(2): 361-7, 1978 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29888

RESUMO

The cytosolic fumarase [EC 4.2.1.2[ of rat liver was bound, after dialysis, to the microsomal membrane in vitro. Binding of the enzyme was dependent on pH, and was facilitated in the pH range below 7.5. The binding reaction was completely inhibited by 0.5 mM fumarate, aurintricarboxylate or colchicine. The bound fumarase was released from the membrane by the substrates, isocitrate, citrate or 2,3-diphosphoglycerate at low concentrations. Desorption of the enzyme by metabolites was also dependent on pH, and was more rapid in the alkaline pH range. The enzyme desorption curves were sigmoidal, and kinetic studies suggested a biphasic cooperative mechanism for the action of the metabolites. The apparent desorption constants (concentrations necessary for 50% desorption of the enzyme) estimated at pH 7.3 for isocitrate, 2,3-diphosphoglycerate, L-malate, oxalacetate, fumarate, citrate, succinate, and KCl were 0.073, 0.074, 0.22, 0.39, 0.56, 2.9, and 19 mM, respectively. The bound fumarase showed little enzymatic activity, and its Km and Vmax values were fivefold and 31%, respectively, of those of the free enzyme.


Assuntos
Fumarato Hidratase/metabolismo , Fígado/enzimologia , Adsorção , Animais , Citosol/enzimologia , Fumarato Hidratase/antagonistas & inibidores , Concentração de Íons de Hidrogênio , Membranas Intracelulares/enzimologia , Cinética , Masculino , Microssomos/metabolismo , Ligação Proteica , Ratos
18.
FEMS Microbiol Lett ; 70(2): 101-6, 1992 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-1587456

RESUMO

A nuclear mutant of Saccharomyces cerevisiae deficient in mitochondrial fumarase has been identified through the in vitro biochemical assay of enzyme activity after visual selection due to an increased acidification ability of its colonies. Cells of the fumarase-deficient mutant fermenting glucose accumulated extracellular fumaric acid. This accumulation was observed only in growing cultures and required functional mitochondrial electron transport from succinate dehydrogenase to oxygen.


Assuntos
Fumarato Hidratase/genética , Fumaratos/metabolismo , Mutação , Saccharomyces cerevisiae/enzimologia , Anaerobiose , Antifúngicos/farmacologia , Cromatografia Líquida de Alta Pressão , Ácidos Graxos Insaturados/farmacologia , Fumarato Hidratase/antagonistas & inibidores , Fumarato Hidratase/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Metacrilatos , Consumo de Oxigênio , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Estrobilurinas
19.
Cancer Chemother Pharmacol ; 29(6): 467-70, 1992.
Artigo em Inglês | MEDLINE | ID: mdl-1314713

RESUMO

The reversibility of cisplatin-protein interactions by the modulating agent WR2721, its active thiol-metabolite WR1065, and the symmetrical disulfide WR33278 was studied using the model compounds (Pt(diethylenetriammine) monofunctionally bound to the sulfur in glutathione (Pt(dien)SG) and Pt(diethylenetriammine) monofunctionally bound to the sulfur in S-methylglutathione (Pt(dien)SMeG). Both model compounds could be quantified by high-performance liquid chromatography (HPLC) with UV detection. The Pt-cysteine-like bond in Pt(dien)SG could not be reversed by any of the WR compounds or by the strong nucleophiles thiosulfate (TS) and diethyldithiocarbamate (DDTC). However, the Pt-methionine-like bond in Pt(dien)SMeG could be reversed by WR1065, although the reversal was slow (k2 = 0.142 M-1 s-1) as compared with that obtained using the modulating agents TS (k2 = 10.1 M-1 s-1) and DDTC (k2 = 3.66 M-1 s-1). WR2721 was hardly able to reverse the Pt-S bond in Pt(dien)SMeG (k2 = 0.00529 M-1 s-1), and WR33278 showed no capacity to do so. The activity of cis-diamminedichloroplatinum(II) (CDDP)-inactivated fumarase was not appreciably restored by any of the WR compounds (16%, 7.7%, and 0 for 20 mM WR1065, WR2721, and WR33278, respectively) in contrast to the strong nucleophile DDTC (61% for 2 mM DDTC). These in vitro studies provide information at the molecular level that may explain why WR2721, in contrast to DDTC, does not provide protection against cisplatin-induced nephrotoxicity when it is given after platinum-containing chemotherapy. The results support the present clinical use of WR2721 prior to the administration of platinum compounds.


Assuntos
Amifostina/farmacologia , Cisplatino/antagonistas & inibidores , Fumarato Hidratase/antagonistas & inibidores , Mercaptoetilaminas/farmacologia , Protetores contra Radiação/farmacologia , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Cisplatino/análogos & derivados , Cisplatino/farmacologia , Ditiocarb/farmacologia , Interações Medicamentosas , Estabilidade de Medicamentos , Fumarato Hidratase/efeitos dos fármacos , Glutationa/análogos & derivados , Glutationa/farmacologia , Cinética , Espectrofotometria Ultravioleta
20.
J Inorg Biochem ; 41(1): 17-24, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1850448

RESUMO

The enzyme fumarase is inhibited by [cis-Pt(NH3)2(H2O)2] (NO3)2. The Pt compound most likely binds at a S-methionine site. Sodium diethyldithiocarbamate (Naddtc) appears to be a powerful regenerator of enzymatic activity. Thiourea is less active, while sodium thiosulfate (STS) is almost inactive in restoring the activity of the enzyme. The regeneration phenomena are based on the dissociation of the Pt-S bonds of the methionine type, and formation of species like [Pt(ddtc)2]. In the model adduct [Pt(dien)GS-Me]2+ Naddtc, thiourea and STS easily break the Pt-S bond of the methionine type. It is concluded that the model system for Naddtc and thiourea does resemble fumarase quite well. S-donor ligands, which may be used as rescue agents in Pt antitumor therapy, are known to suppress nephrotoxicity caused by [cis-PtCl2(NH3)2]. A parallel is drawn between the enzyme reactivation, modeled by fumarase, and the [cis-PtCl2(NH3)2] nephrotoxicity suppression by rescue agents. It is proposed that a Pt-methionine type binding is broken by the rescue agents Naddtc and thiourea, but that the rescue agent STS only inhibits the nephrotoxicity by inactivating unbound Pt species in the cell.


Assuntos
Cisplatino/análogos & derivados , Ditiocarb/farmacologia , Fumarato Hidratase/metabolismo , Tiossulfatos/farmacologia , Tioureia/farmacologia , Animais , Sítios de Ligação , Cisplatino/farmacologia , Fumarato Hidratase/antagonistas & inibidores , Cinética , Ligantes , Modelos Teóricos , Miocárdio/enzimologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA