Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
1.
Acta Biochim Biophys Sin (Shanghai) ; 51(3): 293-300, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30883649

RESUMO

Diabetic cardiomyopathy is an important contributor to morbidity and mortality of diabetic patients by causing heart failure. Interstitial and perivascular fibrosis plays a crucial role in diabetic cardiomyopathy. However, there is a lack of effective specific treatments available for diabetic cardiomyopathy. In the present study, we aim to explore the effects of Liraglutide, a GLP-1 analogue, on diabetic cardiomyopathy in STZ-induced diabetic rats fed with high-fat diet. A total of 60 male Wistar rats were randomly assigned to three groups, i.e. normal group, model group, and Liraglutide group, with 20 rats in each group. Serum levels of TC, TG, LDL-C, NEFA, and hydroxyproline were measured using commercial kits. Cardiac function was evaluated by QRS waves, LVEDd, LVESd, and LVEF. Myocardial fibrosis was measured by immunohistochemistry. Our results demonstrated that chronic administration of Liraglutide decreased the level of blood glucose and significantly alleviated lipid metabolic disturbance compared with the model group. Furthermore, Liraglutide was found to improve the damaged cardiac function. In line with this, we also found that the alleviation of cardiac dysfunction was associated with the decreased fibrosis in diabetic myocardial tissues, which was reflected by the decreased expressions of P4hα-1, COL-1, COL-3, MMP-1, and MMP-9. Our results thus suggest that Liraglutide might have a myocardial protective effect by inhibiting P4hα-1-mediated myocardial fibrosis.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Cardiomiopatias Diabéticas/tratamento farmacológico , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Liraglutida/uso terapêutico , Miocárdio/patologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Animais , Glicemia/análise , Diabetes Mellitus Experimental/complicações , Fibrose , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Estreptozocina
2.
Biochemistry ; 57(13): 2035-2043, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29521097

RESUMO

Protein disulfide isomerase A1 (PDIA1) is an endoplasmic reticulum (ER)-localized thiol-disulfide oxidoreductase that is an important folding catalyst for secretory pathway proteins. PDIA1 contains two active-site domains (a and a'), each containing a Cys-Gly-His-Cys (CGHC) active-site motif. The two active-site domains share 37% sequence identity and function independently to perform disulfide-bond reduction, oxidation, and isomerization. Numerous inhibitors for PDIA1 have been reported, yet the selectivity of these inhibitors toward the a and a' sites is poorly characterized. Here, we identify a potent and selective PDIA1 inhibitor, KSC-34, with 30-fold selectivity for the a site over the a' site. KSC-34 displays time-dependent inhibition of PDIA1 reductase activity in vitro with a kinact/ KI of 9.66 × 103 M-1 s-1 and is selective for PDIA1 over other members of the PDI family, and other cellular cysteine-containing proteins. We provide the first cellular characterization of an a-site selective PDIA1 inhibitor and demonstrate that KSC-34 has minimal sustained effects on the cellular unfolded protein response, indicating that a-site inhibition does not induce global protein folding-associated ER stress. KSC-34 treatment significantly decreases the rate of secretion of a destabilized, amyloidogenic antibody light chain, thereby minimizing pathogenic amyloidogenic extracellular proteins that rely on high PDIA1 activity for proper folding and secretion. Given the poor understanding of the contribution of each PDIA1 active site to the (patho)physiological functions of PDIA1, site selective inhibitors like KSC-34 provide useful tools for delineating the pathological role and therapeutic potential of PDIA1.


Assuntos
Inibidores Enzimáticos/química , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/química , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/química , Dobramento de Proteína , Domínio Catalítico , Humanos , Pró-Colágeno-Prolina Dioxigenase/genética , Isomerases de Dissulfetos de Proteínas/genética
3.
J Pathol ; 241(4): 547-558, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27981571

RESUMO

Prolyl hydroxylase domain-containing proteins (PHDs) regulate the adaptation of cells to hypoxia. Pan-hydroxylase inhibition is protective in experimental colitis, in which PHD1 plays a prominent role. However, it is currently unknown how PHD1 targeting regulates this protection and which cell type(s) are involved. Here, we demonstrated that Phd1 deletion in endothelial and haematopoietic cells (Phd1f/f Tie2:cre) protected mice from dextran sulphate sodium (DSS)-induced colitis, with reduced epithelial erosions, immune cell infiltration, and colonic microvascular dysfunction, whereas the response of Phd2f/+ Tie2:cre and Phd3f/f Tie2:cre mice to DSS was similar to that of their littermate controls. Using bone marrow chimeras and cell-specific cre mice, we demonstrated that ablation of Phd1 in haematopoietic cells but not in endothelial cells was both necessary and sufficient to inhibit experimental colitis. This effect relied, at least in part, on skewing of Phd1-deficient bone marrow-derived macrophages towards an anti-inflammatory M2 phenotype. These cells showed an attenuated nuclear factor-κB-dependent response to lipopolysaccharide (LPS), which in turn diminished endothelial chemokine expression. In addition, Phd1 deficiency in dendritic cells significantly reduced interleukin-1ß production in response to LPS. Taken together, our results further support the development of selective PHD1 inhibitors for ulcerative colitis, and identify haematopoietic cells as their primary target. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Colite Ulcerativa/tratamento farmacológico , Macrófagos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Colite Ulcerativa/imunologia , Colite Ulcerativa/patologia , Colo/efeitos dos fármacos , Colo/patologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Deleção de Genes , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Lipopolissacarídeos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Pró-Colágeno-Prolina Dioxigenase/deficiência , Pró-Colágeno-Prolina Dioxigenase/genética
4.
Kidney Int ; 92(4): 900-908, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28506759

RESUMO

Complement C1q is part of the C1 macromolecular complex that mediates the classical complement activation pathway: a major arm of innate immune defense. C1q is composed of A, B, and C chains that require post-translational prolyl 4-hydroxylation of their N-terminal collagen-like domain to enable the formation of the functional triple helical multimers. The prolyl 4-hydroxylase(s) that hydroxylate C1q have not previously been identified. Recognized prolyl 4-hydroxylases include collagen prolyl-4-hydroxylases (CP4H) and the more recently described prolyl hydroxylase domain (PHD) enzymes that act as oxygen sensors regulating hypoxia-inducible factor (HIF). We show that several small-molecule prolyl hydroxylase inhibitors that activate HIF also potently suppress C1q secretion by human macrophages. However, reducing oxygenation to a level that activates HIF does not compromise C1q hydroxylation. In vitro studies showed that a C1q A chain peptide is not a substrate for PHD2 but is a substrate for CP4H1. Circulating levels of C1q did not differ between wild-type mice or mice with genetic deficits in PHD enzymes, but were reduced by prolyl hydroxylase inhibitors. Thus, C1q is hydroxylated by CP4H, but not the structurally related PHD hydroxylases. Hence, reduction of C1q levels may be an important off-target side effect of small molecule PHD inhibitors developed as treatments for renal anemia.


Assuntos
Complemento C1q/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Macrófagos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Inibidores de Prolil-Hidrolase/farmacologia , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Linhagem Celular , Complemento C1q/análise , Via Clássica do Complemento , Feminino , Humanos , Hidroxilação , Nefropatias/sangue , Nefropatias/tratamento farmacológico , Nefropatias/patologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Inibidores de Prolil-Hidrolase/uso terapêutico , Processamento de Proteína Pós-Traducional
5.
Kidney Int ; 92(6): 1370-1383, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28847650

RESUMO

Hypoxia in the embryo is a frequent cause of intra-uterine growth retardation, low birth weight, and multiple organ defects. In the kidney, this can lead to low nephron endowment, predisposing to chronic kidney disease and arterial hypertension. A key component in cellular adaptation to hypoxia is the hypoxia-inducible factor pathway, which is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3. In the adult kidney, PHD oxygen sensors are differentially expressed in a cell type-dependent manner and control the production of erythropoietin in interstitial cells. However, the role of interstitial cell PHDs in renal development has not been examined. Here we used a genetic approach in mice to interrogate PHD function in FOXD1-expressing stroma during nephrogenesis. We demonstrate that PHD2 and PHD3 are essential for normal kidney development as the combined inactivation of stromal PHD2 and PHD3 resulted in renal failure that was associated with reduced kidney size, decreased numbers of glomeruli, and abnormal postnatal nephron formation. In contrast, nephrogenesis was normal in animals with individual PHD inactivation. We furthermore demonstrate that the defect in nephron formation in PHD2/PHD3 double mutants required intact hypoxia-inducible factor-2 signaling and was dependent on the extent of stromal hypoxia-inducible factor activation. Thus, hypoxia-inducible factor prolyl-4-hydroxylation in renal interstitial cells is critical for normal nephron formation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/fisiologia , Rim/crescimento & desenvolvimento , Pró-Colágeno-Prolina Dioxigenase/fisiologia , Insuficiência Renal/genética , Anemia/sangue , Anemia/tratamento farmacológico , Anemia/etiologia , Animais , Hipóxia Celular/fisiologia , Ensaios Clínicos Fase III como Assunto , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Hidroxilação/fisiologia , Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Rim/citologia , Rim/metabolismo , Nefropatias/complicações , Nefropatias/tratamento farmacológico , Camundongos , Terapia de Alvo Molecular/métodos , Mutação , Tamanho do Órgão/fisiologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , Insuficiência Renal/mortalidade , Insuficiência Renal/patologia , Células Estromais/metabolismo
6.
Am J Respir Cell Mol Biol ; 55(6): 878-888, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27494234

RESUMO

Alveolar epithelial injury and increased alveolar permeability are hallmarks of acute respiratory distress syndrome. Apoptosis of lung epithelial cells via the Fas/Fas ligand (FasL) pathway plays a critical role in alveolar epithelial injury. Activation of hypoxia-inducible factor (HIF)-1 by inhibition of prolyl hydroxylase domain proteins (PHDs) is a possible therapeutic approach to attenuate apoptosis and organ injury. Here, we investigated whether treatment with dimethyloxalylglycine (DMOG), an inhibitor of PHDs, could attenuate Fas/FasL-dependent apoptosis in lung epithelial cells and lung injury. DMOG increased HIF-1α protein expression in vitro in MLE-12 cells, a murine alveolar epithelial cell line. Treatment of MLE-12 cells with DMOG significantly suppressed cell surface expression of Fas and attenuated FasL-induced caspase-3 activation and apoptotic cell death. Inhibition of the HIF-1 pathway by echinomycin or small interfering RNA transfection abolished these antiapoptotic effects of DMOG. Moreover, intraperitoneal injection of DMOG in mice increased HIF-1α expression and decreased Fas expression in lung tissues. DMOG treatment significantly attenuated caspase-3 activation, apoptotic cell death in lung tissue, and the increase in alveolar permeability in mice instilled intratracheally with FasL. In addition, inflammatory responses and histopathological changes were also significantly attenuated by DMOG treatment. In conclusion, inhibition of PHDs protects lung epithelial cells from Fas/FasL-dependent apoptosis through HIF-1 activation and attenuates lung injury in mice.


Assuntos
Apoptose/efeitos dos fármacos , Proteína Ligante Fas/farmacologia , Lesão Pulmonar/enzimologia , Lesão Pulmonar/patologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Aminoácidos Dicarboxílicos/farmacologia , Animais , Caspase 3/metabolismo , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos Endogâmicos C57BL , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Estabilidade Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
7.
Hum Mol Genet ; 22(4): 646-55, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23118353

RESUMO

Protein disulfide isomerase (PDI) is an oxidoreductase assisting oxidative protein folding in the endoplasmic reticulum of all types of cells, including neurons and glia. In neurodegenerative disorders, such as amyotrophic lateral sclerosis (ALS), up-regulation of PDI is an important part of unfolded protein response (UPR) that is thought to represent an adaption reaction and thereby protect the neurons. Importantly, studies on animal models of familial ALS with mutant Cu/Zn superoxide dismutase 1 (SOD1) have shown that the mutant SOD1 in astrocytes or microglia strongly regulates the progression of the disease. Here, we found an early up-regulation of PDI in microglia of transgenic (tg) mutant SOD1 mice, indicating that in addition to neurons, UPR takes place in glial cells in ALS. The observation was supported by the finding that also the expression of a UPR marker GADD34 (growth arrest and DNA damage-inducible protein) was induced in the spinal cord glia of tg mutant SOD1 mice. Because mutant SOD1 can cause sustained activation of NADPH oxidase (NOX), we investigated the role of PDI in UPR-induced NOX activation in microglia. In BV-2 microglia, UPR resulted in NOX activation with increased production of superoxide and increased release of tumor necrosis factor-α. The phenomenon was recapitulated in primary rat microglia, murine macrophages and human monocytes. Importantly, pharmacological inhibition of PDI or its down-regulation by short interfering RNAs prevented NOX activation in microglia and subsequent production of superoxide. Thus, results strongly demonstrate that UPR, caused by protein misfolding, may lead to PDI-dependent NOX activation and contribute to neurotoxicity in neurodegenerative diseases including ALS.


Assuntos
Microglia/enzimologia , NADH NADPH Oxirredutases/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Isomerases de Dissulfetos de Proteínas/metabolismo , Superóxidos/metabolismo , Esclerose Lateral Amiotrófica/enzimologia , Esclerose Lateral Amiotrófica/patologia , Animais , Células do Corno Anterior/enzimologia , Astrócitos/enzimologia , Linhagem Celular , Ativação Enzimática , Indução Enzimática , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Muscimol/análogos & derivados , Muscimol/farmacologia , NADPH Oxidase 1 , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Isomerases de Dissulfetos de Proteínas/genética , Transporte Proteico , Superóxido Dismutase , Superóxido Dismutase-1 , Fator de Necrose Tumoral alfa/metabolismo , Resposta a Proteínas não Dobradas
8.
Bioorg Med Chem ; 23(13): 3081-90, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-26022078

RESUMO

Collagen is the most abundant protein in animals. A variety of indications are associated with the overproduction of collagen, including fibrotic diseases and cancer metastasis. The stability of collagen relies on the posttranslational modification of proline residues to form (2S,4R)-4-hydroxyproline. This modification is catalyzed by collagen prolyl 4-hydroxylases (CP4Hs), which are Fe(II)- and α-ketoglutarate (AKG)-dependent dioxygenases located in the lumen of the endoplasmic reticulum. Human CP4Hs are validated targets for treatment of both fibrotic diseases and metastatic breast cancer. Herein, we report on 2,2'-bipyridinedicarboxylates as inhibitors of a human CP4H. Although most 2,2'-bipyridinedicarboxylates are capable of inhibition via iron sequestration, the 4,5'- and 5,5'-dicarboxylates were found to be potent competitive inhibitors of CP4H, and the 5,5'-dicarboxylate was selective in its inhibitory activity. Our findings clarify a strategy for developing CP4H inhibitors of clinical utility.


Assuntos
Ácidos Carboxílicos/química , Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Quelantes de Ferro/química , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Inibidores de Prolil-Hidrolase/química , Piridinas/química , Animais , Ligação Competitiva , Ácidos Carboxílicos/síntese química , Colágeno/antagonistas & inibidores , Colágeno/biossíntese , Relação Dose-Resposta a Droga , Ensaios Enzimáticos , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/química , Ferro/metabolismo , Quelantes de Ferro/síntese química , Isoenzimas/antagonistas & inibidores , Isoenzimas/química , Cinética , Pró-Colágeno-Prolina Dioxigenase/química , Inibidores de Prolil-Hidrolase/síntese química , Piridinas/síntese química , Proteínas Recombinantes/química
9.
Blood ; 120(16): 3336-44, 2012 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22955912

RESUMO

An endoplasmic reticulum transmembrane prolyl 4-hydroxylase (P4H-TM) is able to hydroxylate the α subunit of the hypoxia-inducible factor (HIF) in vitro and in cultured cells, but nothing is known about its roles in mammalian erythropoiesis. We studied such roles here by administering a HIF-P4H inhibitor, FG-4497, to P4h-tm(-/-) mice. This caused larger increases in serum Epo concentration and kidney but not liver Hif-1α and Hif-2α protein and Epo mRNA levels than in wild-type mice, while the liver Hepcidin mRNA level was lower in the P4h-tm(-/-) mice than in the wild-type. Similar, but not identical, differences were also seen between FG-4497-treated Hif-p4h-2 hypomorphic (Hif-p4h-2(gt/gt)) and Hif-p4h-3(-/-) mice versus wild-type mice. FG-4497 administration increased hemoglobin and hematocrit values similarly in the P4h-tm(-/-) and wild-type mice, but caused higher increases in both values in the Hif-p4h-2(gt/gt) mice and in hematocrit value in the Hif-p4h-3(-/-) mice than in the wild-type. Hif-p4h-2(gt/gt)/P4h-tm(-/-) double gene-modified mice nevertheless had increased hemoglobin and hematocrit values without any FG-4497 administration, although no such abnormalities were seen in the Hif-p4h-2(gt/gt) or P4h-tm(-/-) mice. Our data thus indicate that P4H-TM plays a role in the regulation of EPO production, hepcidin expression, and erythropoiesis.


Assuntos
Eritropoese/fisiologia , Eritropoetina/sangue , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Pró-Colágeno-Prolina Dioxigenase/fisiologia , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Western Blotting , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Hematócrito , Hemoglobinas/metabolismo , Hepcidinas , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
BMC Cancer ; 14: 1, 2014 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-24383403

RESUMO

BACKGROUND: Increased collagen deposition provides physical and biochemical signals to support tumor growth and invasion during breast cancer development. Therefore, inhibition of collagen synthesis and deposition has been considered a strategy to suppress breast cancer progression. Collagen prolyl-4-hydroxylase α subunit 2 (P4HA2), an enzyme hydroxylating proline residues in -X-Pro-Gly- sequences, is a potential therapeutic target for the disorders associated with increased collagen deposition. However, expression and function of P4HA2 in breast cancer progression are not well investigated. METHODS: Gene co-expression analysis was performed in the published microarray datasets to identify potential regulators of collagen I, III, and IV in human breast cancer tissue. Expression of P4HA2 was silenced by shRNAs, and its activity was inhibited by 1, 4-DPCA, a prolyl-4-hydroxylase inhibitor. Three-dimensional culture assay was used to analyze roles of P4HA2 in regulating malignant phenotypes of breast cancer cells. Reduced deposition of collagen I and IV was detected by Western blotting and immunofluorescence. Control and P4HA2-silenced breast cancer cells were injected into fat pad and tail vein of SCID mice to examine effect of P4HA2 on tumor growth and lung metastasis. RESULTS: Using gene co-expression analysis, we showed that P4HA2 was associated with expression of Col1A1, Col3A1, and Col4A1 during breast cancer development and progression. P4HA2 mRNA levels were significantly upregulated in breast cancer compared to normal mammary tissue. Increased mRNA levels of P4HA2 correlated with poor clinical outcome in breast cancer patients, which is independent of estrogen receptor status. Silencing P4HA2 expression or treatment with the P4HA inhibitor significantly inhibited cell proliferation and suppressed aggressive phenotypes of breast cancer cells in 3D culture, accompanied by reduced deposition of collagen I and IV. We also found that knockdown of P4HA2 inhibited mammary tumor growth and metastasis to lungs in xenograft models. CONCLUSION: These results suggest the critical role of P4HA2 in breast cancer progression and identify P4HA2 as a potential therapeutic target and biomarker for breast cancer progression.


Assuntos
Neoplasias da Mama/enzimologia , Colágeno/metabolismo , Neoplasias Pulmonares/enzimologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolil Hidroxilases/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colágeno/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Colágeno Tipo III/metabolismo , Colágeno Tipo IV/metabolismo , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Feminino , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Camundongos , Camundongos SCID , Invasividade Neoplásica , Fenótipo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , Prognóstico , Prolil Hidroxilases/genética , Interferência de RNA , RNA Mensageiro/metabolismo , Fatores de Tempo , Transfecção , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Am Soc Nephrol ; 24(8): 1234-41, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23661806

RESUMO

The identity of the peritubular population of cells with mesenchymal phenotype thought responsible for producing erythropoietin in humans remains unclear. Here, renal CD133(+)/CD73(+) progenitor cells, isolated from the human renal inner medulla and described as a population of mesenchymal progenitors, released erythropoietin under hypoxic conditions. CD133(-) cells did not synthesize erythropoietin, and CD133(+) progenitor cells stopped producing erythropoietin when they differentiated and acquired an epithelial phenotype. Inhibition of prolyl hydroxylases, using either dimethyloxalylglycine or a small hairpin RNA against prolyl hydroxylase-2, increased both hypoxia-inducible factor-2α (HIF-2α) expression and erythropoietin transcription. Moreover, under hypoxic conditions, inhibition of prolyl hydroxylase significantly increased erythropoietin release by CD133(+) progenitors. Finally, blockade of HIF-2α impaired erythropoietin synthesis by CD133(+) progenitors. Taken together, these results suggest that it is the renal CD133(+) progenitor cells that synthesize and release erythropoietin under hypoxia, via the prolyl hydroxylase-HIF-2α axis, in the human kidney. In addition, this study provides rationale for the therapeutic use of prolyl hydroxylase inhibitors in the setting of acute or chronic renal injury.


Assuntos
Eritropoetina/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Medula Renal/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , 5'-Nucleotidase/análise , Antígeno AC133 , Antígenos CD/análise , Proteínas Ligadas por GPI/análise , Glicoproteínas/análise , Humanos , Peptídeos/análise , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Células-Tronco/metabolismo
12.
J Cell Biochem ; 114(4): 864-73, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23097160

RESUMO

Hypoxia inducible factor 1α (HIF-1α), an essential transcriptional factor, is negatively regulated by two different types of oxygen and Fe(2+) -dependent HIF hydroxylases, proline hydroxylase (PHD) and factor inhibiting HIF (FIH), under normoxia. Iron chelators have therefore been used for inducing HIF-1α expression by inhibiting the hydroxylases. In this study, the iron chelators displayed differential effects for PHD and FIH in cells depending on their iron specificity and membrane permeability rather than their in vitro potencies. The membrane permeability of the strict Fe(2+) -chelator potentially inhibited both hydroxylases, whereas the membrane impermeable one showed no inhibitory effect in cells. In contrast, the depletion of the extracellular Fe(3+) ion was mainly correlated to PHD inhibition, and the membrane permeable one elicited low efficacy for both enzymes in cells. The 3'-hydroxyl group of quercetin, a natural flavonoid, was critical for inhibition of intracellular hydroxylases. Since the 3'-methylation of quercetin is induced by catechol-O-methyl transferase, the enzyme may regulate the intracellular activity of quercetin. These data suggest that the multiple factors of iron-chelators may be responsible for regulating the intracellular activity HIF hydroxylases.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Quelantes de Ferro/farmacologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Animais , Anticorpos Monoclonais Murinos/metabolismo , Permeabilidade da Membrana Celular , Clonagem Molecular , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Compostos Férricos/metabolismo , Células HeLa , Humanos , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia , Ferro/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Fenantrolinas/farmacologia , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , Ligação Proteica , Quercetina/análogos & derivados , Quercetina/farmacologia , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transcrição Gênica
13.
Hippocampus ; 23(10): 861-72, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23674383

RESUMO

Hypoxia-inducible factors (HIFs) are key transcriptional regulators that play a major role in oxygen homeostasis. HIF activity is tightly regulated by oxygen-dependent hydroxylases, which additionally require iron and 2-oxoglutarate as cofactors. Inhibition of these enzymes has become a novel target to modulate the hypoxic response for therapeutic benefit. Inhibition of prolyl-4-hydroxylase domains (PHDs) have been shown to delay neuronal cell death and protect against ischemic injury in the hippocampus. In this study we have examined the effects of prolyl hydroxylase inhibition on synaptic transmission and plasticity in the hippocampus. Field excitatory postsynaptic potentials (fEPSPs) and excitatory postsynaptic currents (EPSCs) were elicited by stimulation of the Schaffer collateral pathway in the CA1 region of the hippocampus. Treatment of rat hippocampal slices with low concentrations (10 µM) of the iron chelator deferosoxamine (DFO) or the 2-oxoglutarate analogue dimethyloxalyl glycine (DMOG) had no effect on fEPSP. In contrast, application of 1 mM DMOG resulted in a significant decrease in fEPSP slope. Antagonism of the NMDA receptor attenuated the effects of DMOG on baseline synaptic signalling. In rat hippocampal slices pretreated with DMOG and DFO the induction of long-term potentiation (LTP) by tetanic stimulation was strongly impaired. Similarly, neuronal knockout of the single PHD family member PHD2 prevented murine hippocampal LTP. Preconditioning of PHD2 deficient hippocampi with either DMOG, DFO, or the PHD specific inhibitor JNJ-42041935, did not further decrease LTP suggesting that DMOG and DFO influences synaptic plasticity primarily by inhibiting PHDs rather than unspecific effects. These findings provide striking evidence for a modulatory role of PHD proteins on synaptic plasticity in the hippocampus.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/enzimologia , Potenciação de Longa Duração/fisiologia , Pró-Colágeno-Prolina Dioxigenase/fisiologia , Aminoácidos Dicarboxílicos/farmacologia , Animais , Região CA1 Hipocampal/efeitos dos fármacos , Região CA1 Hipocampal/enzimologia , Desferroxamina/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp/instrumentação , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Ratos , Ratos Wistar
14.
Am J Transplant ; 13(3): 600-10, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23331861

RESUMO

Ischemia-reperfusion injury (IRI) induces hypoxia-inducible factor-1 (HIF-1) in the myocardium, but the consequences remain elusive. We investigated HIF-1 activation during cold and warm ischemia and IRI in rat hearts and cardiac syngrafts. We also tested the effect of HIF-α stabilizing prolyl hydroxylase inhibitor (FG-4497) on IRI or allograft survival. Ex vivo ischemia of the heart increased HIF-1α expression in a time- and temperature-dependent fashion. Immunohistochemistry localized HIF-1α to all cardiac cell types. After reperfusion, HIF-1α immunoreactivity persisted in smooth muscle cells and cardiomyocytes in the areas with IRI. This was accompanied with a transient induction of protective HIF-1 downstream genes. Donor FG-4497 pretreatment for 4 h enhanced IRI in cardiac allografts as evidenced by an increase in cardiac troponin T release, cardiomyocyte apoptosis, and activation of innate immunity. Recipient FG-4497 pretreatment for 4 h decreased infiltration of ED1(+) macrophages, and mildly improved the long-term allograft survival. In syngrafts donor FG-4497 pretreatment increased activation of innate immunity, but did not induce myocardial damage. We conclude that the HIF-1 pathway is activated in heart transplants. We suggest that pharmacological HIF-α preconditioning of cardiac allografts donors would not lead to clinical benefit, while in recipients it may result in antiinflammatory effects and prolonged allograft survival.


Assuntos
Inibidores Enzimáticos/farmacologia , Transplante de Coração , Coração/fisiopatologia , Fator 1 Induzível por Hipóxia/metabolismo , Precondicionamento Isquêmico , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Inflamação/diagnóstico , Inflamação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos WF , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transplante Homólogo
15.
Biochem Biophys Res Commun ; 433(1): 40-6, 2013 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-23454121

RESUMO

Thioredoxin interacting protein (TXNIP), first identified as an inhibitor of thioredoxin, is also a tumor suppressor as well as an inhibitor of lipogenesis. TXNIP is known to be transcriptionally regulated in response to nutrients such as glucose and stress signals, including endoplasmic reticulum stress and lactic acidosis. In this study, we characterized the transcriptional regulation of TXNIP in response to hypoxia. Using a hepatocellular carcinoma cell line, we have found that TXNIP mRNA expression is regulated in a biphasic manner in hypoxia whereby TXNIP expression showed an initial rapid decrease, followed by an increase under prolonged hypoxia. Interestingly, we have shown that TXNIP induction in prolonged hypoxia is independent of the Hypoxia-Inducible Factor (HIF) transcription factor. The effect of hypoxia on TXNIP expression is mediated via the inhibition of the 4E-BP1/eIF4E axis of mechanistic target of rapamycin (mTORC1). Thus, we found that inhibiting mTORC1-dependent 4E-BP1 phosphorylation mimics the effect of hypoxia on TXNIP expression. Furthermore, overexpressing eIF4E prevents the induction of TXNIP in hypoxia. Our results suggest that mTORC1 may be an important regulator of hypoxia-dependent gene expression.


Assuntos
Proteínas de Transporte/genética , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Tiorredoxinas/genética , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Animais , Translocador Nuclear Receptor Aril Hidrocarboneto/antagonistas & inibidores , Translocador Nuclear Receptor Aril Hidrocarboneto/genética , Translocador Nuclear Receptor Aril Hidrocarboneto/metabolismo , Sequência de Bases , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/biossíntese , Proteínas de Ciclo Celular , Inibidores Enzimáticos/farmacologia , Fatores de Iniciação em Eucariotos , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células Hep G2 , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Células NIH 3T3 , Fosfoproteínas/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Tiorredoxinas/biossíntese , Transcrição Gênica
16.
J Pharmacol Exp Ther ; 345(1): 25-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23388095

RESUMO

Storage protocols of vascular grafts need further improvement against ischemia-reperfusion (IR) injury. Hypoxia elicits a variety of complex cellular responses by altering the activity of many signaling pathways, such as the oxygen-dependent prolyl-hyroxylase domain-containing enzyme (PHD). Reduction of PHD activity during hypoxia leads to stabilization and accumulation of hypoxia inducible factor (HIF) 1α. We examined the effects of PHD inhibiton by dimethyloxalylglycine on the vasomotor responses of isolated rat aorta and aortic vascular smooth muscle cells (VSMCs) in a model of cold ischemia/warm reperfusion. Aortic segments underwent 24 hours of cold ischemic preservation in saline or DMOG (dimethyloxalylglycine)-supplemented saline solution. We investigated endothelium-dependent and -independent vasorelaxations. To simulate IR injury, hypochlorite (NaOCl) was added during warm reperfusion. VSMCs were incubated in NaCl or DMOG solution at 4°C for 24 hours after the medium was changed for a supplied standard medium at 37°C for 6 hours. Apoptosis was assessed using the TUNEL method. Gene expression analysis was performed using quantitative real-time polymerase chain reaction. Cold ischemic preservation and NaOCl induced severe endothelial dysfunction, which was significantly improved by DMOG supplementation (maximal relaxation of aortic segments to acetylcholine: control 95% ± 1% versus NaOCl 44% ± 4% versus DMOG 68% ± 5%). Number of TUNEL-positive cell nuclei was significantly higher in the NaOCl group, and DMOG treatment significantly decreased apoptosis. Inducible heme-oxygenase 1 mRNA expressions were significantly higher in the DMOG group. Pharmacological modulation of oxygen sensing system by DMOG in an in vitro model of vascular IR effectively preserved endothelial function. Inhibition of PHDs could therefore be a new therapeutic avenue for protecting endothelium and vascular muscle cells against IR injury.


Assuntos
Aorta/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Traumatismo por Reperfusão/prevenção & controle , Vasodilatação/efeitos dos fármacos , Aminoácidos Dicarboxílicos/farmacologia , Animais , Aorta/enzimologia , Aorta/patologia , Apoptose/efeitos dos fármacos , Técnicas de Cultura de Células , Modelos Animais de Doenças , Endotélio Vascular/enzimologia , Endotélio Vascular/patologia , Inibidores Enzimáticos/farmacologia , Heme Oxigenase-1/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Marcação In Situ das Extremidades Cortadas , Contração Isométrica/efeitos dos fármacos , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/patologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/patologia
17.
Blood ; 117(12): 3435-44, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21177435

RESUMO

With the aim of finding small molecules that stimulate erythropoiesis earlier than erythropoietin and that enhance erythroid colony-forming unit (CFU-E) production, we studied the mechanism by which glucocorticoids increase CFU-E formation. Using erythroid burst-forming unit (BFU-E) and CFU-E progenitors purified by a new technique, we demonstrate that glucocorticoids stimulate the earliest (BFU-E) progenitors to undergo limited self-renewal, which increases formation of CFU-E cells > 20-fold. Interestingly, glucocorticoids induce expression of genes in BFU-E cells that contain promoter regions highly enriched for hypoxia-induced factor 1α (HIF1α) binding sites. This suggests activation of HIF1α may enhance or replace the effect of glucocorticoids on BFU-E self-renewal. Indeed, HIF1α activation by a prolyl hydroxylase inhibitor (PHI) synergizes with glucocorticoids and enhances production of CFU-Es 170-fold. Because PHIs are able to increase erythroblast production at very low concentrations of glucocorticoids, PHI-induced stimulation of BFU-E progenitors thus represents a conceptually new therapeutic window for treating erythropoietin-resistant anemia.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/fisiologia , Glucocorticoides/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/agonistas , Aminoácidos Dicarboxílicos/farmacologia , Animais , Sítios de Ligação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Separação Celular , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feto/citologia , Citometria de Fluxo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Fígado/citologia , Fígado/embriologia , Camundongos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética
18.
EMBO Rep ; 12(5): 463-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21460794

RESUMO

Mutations in isocitrate dehydrogenases (IDHs) have a gain-of-function effect leading to R(-)-2-hydroxyglutarate (R-2HG) accumulation. By using biochemical, structural and cellular assays, we show that either or both R- and S-2HG inhibit 2-oxoglutarate (2OG)-dependent oxygenases with varying potencies. Half-maximal inhibitory concentration (IC(50)) values for the R-form of 2HG varied from approximately 25 µM for the histone N(ɛ)-lysine demethylase JMJD2A to more than 5 mM for the hypoxia-inducible factor (HIF) prolyl hydroxylase. The results indicate that candidate oncogenic pathways in IDH-associated malignancy should include those that are regulated by other 2OG oxygenases than HIF hydroxylases, in particular those involving the regulation of histone methylation.


Assuntos
Glutaratos/metabolismo , Histona Desmetilases/antagonistas & inibidores , Isocitrato Desidrogenase/genética , Modelos Moleculares , Neoplasias/enzimologia , Transdução de Sinais/fisiologia , Linhagem Celular Tumoral , Cristalografia , Humanos , Concentração Inibidora 50 , Isocitrato Desidrogenase/metabolismo , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Histona Desmetilases com o Domínio Jumonji/química , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Oxigenases de Função Mista , Mutação/genética , Neoplasias/genética , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/química
19.
Org Biomol Chem ; 11(5): 732-745, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23151668

RESUMO

Inhibition of the hypoxia-inducible factor (HIF) prolyl hydroxylases (PHD or EGLN enzymes) is of interest for the treatment of anemia and ischemia-related diseases. Most PHD inhibitors work by binding to the single ferrous ion and competing with 2-oxoglutarate (2OG) co-substrate for binding at the PHD active site. Non-specific iron chelators also inhibit the PHDs, both in vitro and in cells. We report the identification of dual action PHD inhibitors, which bind to the active site iron and also induce the binding of a second iron ion at the active site. Following analysis of small-molecule iron complexes and application of non-denaturing protein mass spectrometry to assess PHD2·iron·inhibitor stoichiometry, selected diacylhydrazines were identified as PHD2 inhibitors that induce the binding of a second iron ion. Some compounds were shown to inhibit the HIF hydroxylases in human hepatoma and renal carcinoma cell lines.


Assuntos
Hidrazinas/química , Hidrazinas/farmacologia , Ferro/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Domínio Catalítico , Linhagem Celular Tumoral , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Simulação de Acoplamento Molecular , Pró-Colágeno-Prolina Dioxigenase/química , Ligação Proteica/efeitos dos fármacos , Espectrometria de Massas por Ionização por Electrospray
20.
Cancer Cell ; 7(1): 77-85, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15652751

RESUMO

Several mitochondrial proteins are tumor suppressors. These include succinate dehydrogenase (SDH) and fumarate hydratase, both enzymes of the tricarboxylic acid (TCA) cycle. However, to date, the mechanisms by which defects in the TCA cycle contribute to tumor formation have not been elucidated. Here we describe a mitochondrion-to-cytosol signaling pathway that links mitochondrial dysfunction to oncogenic events: succinate, which accumulates as a result of SDH inhibition, inhibits HIF-alpha prolyl hydroxylases in the cytosol, leading to stabilization and activation of HIF-1alpha. These results suggest a mechanistic link between SDH mutations and HIF-1alpha induction, providing an explanation for the highly vascular tumors that develop in the absence of VHL mutations.


Assuntos
Ciclo do Ácido Cítrico/fisiologia , Oncogenes , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Ácido Succínico/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Transformação Celular Neoplásica , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mitocôndrias/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Succinato Desidrogenase/antagonistas & inibidores , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA