Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Impot Res ; 30(3): 108-116, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29736011

RESUMO

cGMP-independent nitric oxide (NO) signaling occurs via S-nitrosylation. We evaluated whether aberrant S-nitrosylation operates in the penis under conditions of cavernous nerve injury and targets proteins involved in regulating erectile function. Adult male Sprague-Dawley rats underwent bilateral cavernous nerve crush injury (BCNI) or sham surgery. Rats were given a denitrosylation agent N-acetylcysteine (NAC, 300 mg/kg/day) or vehicle in drinking water starting 2 days before BCNI and continuing for 2 weeks following surgery. After assessment of erectile function (intracavernous pressure), penes were collected for measurements of S-nitrosylation by Saville-Griess and TMT-switch assays and PKG-I function by immunoblotting of phospho (P)-VASP-Ser-239. Erectile function was decreased (P < 0.05) after BCNI, and it was preserved (P < 0.05) by NAC treatment. Total S-nitrosothiols and total S-nitrosylated proteins were increased (P < 0.05) after BCNI, and these were partially prevented by NAC treatment. S-nitrosylation of sGC was increased (P < 0.05) after BCNI, and it was prevented (P < 0.05) by NAC treatment. S-nitrosylation of eNOS was increased (P < 0.05) after BCNI, and showed a trend towards decrease by NAC treatment. Protein expression of P-VASP-Ser-239 was decreased (P < 0.05) after BCNI, and showed a trend towards increase by NAC treatment. In conclusion, erectile dysfunction following BCNI is mediated in part by S-nitrosylation of eNOS and its downstream signaling mediator GC, while denitrosylation protects erectile function by preserving the NO/cGMP signaling pathway.


Assuntos
Disfunção Erétil/etiologia , Óxido Nítrico Sintase Tipo III/metabolismo , Pênis/inervação , Transdução de Sinais/fisiologia , Acetilcisteína/farmacologia , Animais , GMP Cíclico/metabolismo , Masculino , Compressão Nervosa , Óxido Nítrico/metabolismo , Ereção Peniana/efeitos dos fármacos , Ereção Peniana/fisiologia , Ratos , Ratos Sprague-Dawley
2.
Hum Mutat ; 36(2): 260-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25451944

RESUMO

The greatest interpretive challenge of modern medicine may be to functionally annotate the vast variation of human genomes. Demonstrating a proposed approach, we created a library of BRCA2 exon 27 shotgun-mutant plasmids including solitary and multiplex mutations to generate human knockin clones using homologous recombination. This 55-mutation, 13-clone syngeneic variance library (SyVaL) comprised severely affected clones having early-stop nonsense mutations, functionally hypomorphic clones having multiple missense mutations emphasizing the potential to identify and assess hypomorphic mutations in novel proteomic and epidemiologic studies, and neutral clones having multiple missense mutations. Efficient coverage of nonessential amino acids was provided by mutation multiplexing. Severe mutations were distinguished from hypomorphic or neutral changes by chemosensitivity assays (hypersensitivity to mitomycin C and acetaldehyde), by analysis of RAD51 focus formation, and by mitotic multipolarity. A multiplex unbiased approach of generating all-human SyVaLs in medically important genes, with random mutations in native genes, would provide databases of variants that could be functionally annotated without concerns arising from exogenous cDNA constructs or interspecies interactions, as a basis for subsequent proteomic domain mapping or clinical calibration if desired. Such gene-irrelevant approaches could be scaled up for multiple genes of clinical interest, providing distributable cellular libraries linked to public-shared functional databases.


Assuntos
Proteína BRCA2/genética , Anotação de Sequência Molecular , Substituição de Aminoácidos , Linhagem Celular , Bases de Dados Genéticas , Biblioteca Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Hemizigoto , Recombinação Homóloga , Humanos , Mitose , Rad51 Recombinase/genética
3.
Circ Res ; 115(4): 450-9, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24903103

RESUMO

RATIONALE: Increased arginase activity contributes to endothelial dysfunction by competition for l-arginine substrate and reciprocal regulation of nitric oxide synthase (NOS). The rapid increase in arginase activity in human aortic endothelial cells exposed to oxidized low-density lipoprotein (OxLDL) is consistent with post-translational modification or subcellular trafficking. OBJECTIVE: To test the hypotheses that OxLDL triggers reverse translocation of mitochondrial arginase 2 (Arg2) to cytosol and Arg2 activation, and that this process is dependent on mitochondrial processing peptidase, lectin-like OxLDL receptor-1 receptor, and rho kinase. METHODS AND RESULTS: OxLDL-triggered translocation of Arg2 from mitochondria to cytosol in human aortic endothelial cells and in murine aortic intima with a concomitant rise in arginase activity. All of these changes were abolished by inhibition of mitochondrial processing peptidase or by its siRNA-mediated knockdown. Rho kinase inhibition and the absence of the lectin-like OxLDL receptor-1 in knockout mice also ablated translocation. Aminoterminal sequencing of Arg2 revealed 2 candidate mitochondrial targeting sequences, and deletion of either of these confined Arg2 to the cytoplasm. Inhibitors of mitochondrial processing peptidase or lectin-like OxLDL receptor-1 knockout attenuated OxLDL-mediated decrements in endothelial-specific NO production and increases in superoxide generation. Finally, Arg2(-/-) mice bred on an ApoE(-/-) background showed reduced plaque load, reduced reactive oxygen species production, enhanced NO, and improved endothelial function when compared with ApoE(-/-) controls. CONCLUSIONS: These data demonstrate dual distribution of Arg2, a protein with an unambiguous mitochondrial targeting sequence, in mammalian cells, and its reverse translocation to cytoplasm by alterations in the extracellular milieu. This novel molecular mechanism drives OxLDL-mediated arginase activation, endothelial NOS uncoupling, endothelial dysfunction, and atherogenesis.


Assuntos
Aorta/enzimologia , Arginase/metabolismo , Células Endoteliais/enzimologia , Lipoproteínas LDL/metabolismo , Metaloendopeptidases/metabolismo , Mitocôndrias/enzimologia , Quinases Associadas a rho/metabolismo , Sequência de Aminoácidos , Animais , Aorta/efeitos dos fármacos , Aorta/patologia , Aorta/fisiopatologia , Doenças da Aorta/enzimologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Doenças da Aorta/prevenção & controle , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Arginase/genética , Aterosclerose/enzimologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Células Cultivadas , Citosol/enzimologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Humanos , Masculino , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Dados de Sequência Molecular , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico , Interferência de RNA , Receptores Depuradores Classe E/deficiência , Receptores Depuradores Classe E/genética , Transdução de Sinais , Fatores de Tempo , Transfecção , Quinases Associadas a rho/antagonistas & inibidores , Peptidase de Processamento Mitocondrial
4.
Am J Pathol ; 184(1): 260-70, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24200853

RESUMO

Large-magnitude numerical distinctions (>10-fold) among drug responses of genetically contrasting cancers were crucial for guiding the development of some targeted therapies. Similar strategies brought epidemiological clues and prevention goals for genetic diseases. Such numerical guides, however, were incomplete or low magnitude for Fanconi anemia pathway (FANC) gene mutations relevant to cancer in FANC-mutation carriers (heterozygotes). We generated a four-gene FANC-null cancer panel, including the engineering of new PALB2/FANCN-null cancer cells by homologous recombination. A characteristic matching of FANCC-null, FANCG-null, BRCA2/FANCD1-null, and PALB2/FANCN-null phenotypes was confirmed by uniform tumor regression on single-dose cross-linker therapy in mice and by shared chemical hypersensitivities to various inter-strand cross-linking agents and γ-radiation in vitro. Some compounds, however, had contrasting magnitudes of sensitivity; a strikingly high (19- to 22-fold) hypersensitivity was seen among PALB2-null and BRCA2-null cells for the ethanol metabolite, acetaldehyde, associated with widespread chromosomal breakage at a concentration not producing breaks in parental cells. Because FANC-defective cancer cells can share or differ in their chemical sensitivities, patterns of selective hypersensitivity hold implications for the evolutionary understanding of this pathway. Clinical decisions for cancer-relevant prevention and management of FANC-mutation carriers could be modified by expanded studies of high-magnitude sensitivities.


Assuntos
Acetaldeído/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas de Grupos de Complementação da Anemia de Fanconi/genética , Animais , Western Blotting , Linhagem Celular Tumoral , Anemia de Fanconi/genética , Humanos , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Transfusion ; 54(2): 434-44, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23781865

RESUMO

BACKGROUND: Red blood cell (RBC) deformation is critical for microvascular perfusion and oxygen delivery to tissues. Abnormalities in RBC deformability have been observed in aging, sickle cell disease, diabetes, and preeclampsia. Although nitric oxide (NO) prevents decreases in RBC deformability, the underlying mechanism is unknown. STUDY DESIGN AND METHODS: As an experimental model, we used ionophore A23187-mediated calcium influx in RBCs to reduce their deformability and investigated the role of NO donor sodium nitroprusside (SNP) and KCa3.1 (Gardos) channel blockers on RBC deformability (measured as elongation index [EI] by microfluidic ektacytometry). RBC intracellular Ca(2+) and extracellular K(+) were measured by inductively coupled plasma mass spectrometry and potassium ion selective electrode, respectively. RESULTS: SNP treatment of RBCs blocked the Ca(2+) (approx. 10 µmol/L)-induced decrease in RBC deformability (EI 0.34 ± 0.02 vs. 0.09 ± 0.01, control vs. Ca(2+) loaded, p < 0.001; and EI 0.37 ± 0.02 vs. 0.30 ± 0.01, SNP vs. SNP plus Ca(2+) loaded) as well as Ca(2+) influx and K(+) efflux. The SNP effect was similar to that observed after pharmacologic blockade of the KCa3.1 channel (with charybdotoxin or extracellular medium containing isotonic K(+) concentration). In RBCs from KCa3.1(-/-) mice, 10 µmol/L Ca(2+) loading did not decrease cellular deformability. A preliminary attempt to address the molecular mechanism of SNP protection suggests the involvement of cell surface thiols. CONCLUSION: Our results suggest that nitroprusside treatment of RBCs may protect them from intracellular calcium increase-mediated stiffness, which may occur during microvascular perfusion in diseased states, as well as during RBC storage.


Assuntos
Calcimicina/farmacologia , Cálcio/metabolismo , Deformação Eritrocítica/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Nitroprussiato/farmacologia , Animais , Doadores de Sangue , Ionóforos de Cálcio/farmacologia , Charibdotoxina/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Ácido Iodoacético/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurotoxinas/farmacologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Potássio/farmacologia
6.
Food Chem Toxicol ; 55: 557-67, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23402862

RESUMO

Population differences in age-related diseases and cancer could stem from differences in diet. To characterize DNA strand-breaking activities in selected foods/beverages, flavorings, and some of their constituent chemicals, we used p53R cells, a cellular assay sensitive to such breaks. Substances testing positive included reference chemicals: quinacrine (peak response, 51×) and etoposide (33×); flavonoids: EGCG (19×), curcumin (12×), apigenin (9×), and quercetin (7×); beverages: chamomile (11×), green (21×), and black tea (26×) and coffee (3-29×); and liquid smoke (4-28×). Damage occurred at dietary concentrations: etoposide near 5µg/ml produced responses similar to a 1:1000 dilution of liquid smoke, a 1:20 dilution of coffee, and a 1:5 dilution of tea. Pyrogallol-related chemicals and tannins are present in dietary sources and individually produced strong activity: pyrogallol (30×), 3-methoxycatechol (25×), gallic acid (21×), and 1,2,4-benzenetriol (21×). From structure-activity relationships, high activities depended on specific orientations of hydroxyls on the benzene ring. Responses accompanied cellular signals characteristic of DNA breaks such as H2AX phosphorylation. Breaks were also directly detected by comet assay. Cellular toxicological effects of foods and flavorings could guide epidemiologic and experimental studies of potential disease risks from DNA strand-breaking chemicals in diets.


Assuntos
Dano ao DNA , Aromatizantes/toxicidade , Análise de Alimentos , Linhagem Celular , Humanos
7.
Circ Res ; 109(11): 1259-68, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21980127

RESUMO

RATIONALE: Nitric oxide, the classic endothelium-derived relaxing factor (EDRF), acts through cyclic GMP and calcium without notably affecting membrane potential. A major component of EDRF activity derives from hyperpolarization and is termed endothelium-derived hyperpolarizing factor (EDHF). Hydrogen sulfide (H(2)S) is a prominent EDRF, since mice lacking its biosynthetic enzyme, cystathionine γ-lyase (CSE), display pronounced hypertension with deficient vasorelaxant responses to acetylcholine. OBJECTIVE: The purpose of this study was to determine if H(2)S is a major physiological EDHF. METHODS AND RESULTS: We now show that H(2)S is a major EDHF because in blood vessels of CSE-deleted mice, hyperpolarization is virtually abolished. H(2)S acts by covalently modifying (sulfhydrating) the ATP-sensitive potassium channel, as mutating the site of sulfhydration prevents H(2)S-elicited hyperpolarization. The endothelial intermediate conductance (IK(Ca)) and small conductance (SK(Ca)) potassium channels mediate in part the effects of H(2)S, as selective IK(Ca) and SK(Ca) channel inhibitors, charybdotoxin and apamin, inhibit glibenclamide-insensitive, H(2)S-induced vasorelaxation. CONCLUSIONS: H(2)S is a major EDHF that causes vascular endothelial and smooth muscle cell hyperpolarization and vasorelaxation by activating the ATP-sensitive, intermediate conductance and small conductance potassium channels through cysteine S-sulfhydration. Because EDHF activity is a principal determinant of vasorelaxation in numerous vascular beds, drugs influencing H(2)S biosynthesis offer therapeutic potential.


Assuntos
Endotélio Vascular/metabolismo , Sulfeto de Hidrogênio/metabolismo , Canais KATP/metabolismo , Vasodilatação/fisiologia , Acetilcolina/farmacologia , Animais , Aorta/citologia , Aorta/metabolismo , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Charibdotoxina/farmacologia , Cistationina gama-Liase/deficiência , Cistationina gama-Liase/genética , Fatores Relaxantes Dependentes do Endotélio/metabolismo , Feminino , Glibureto/farmacologia , Hipertensão/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Artérias Mesentéricas/lesões , Artérias Mesentéricas/metabolismo , Artérias Mesentéricas/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fenilefrina/farmacologia , Cloreto de Potássio/farmacologia , Ratos , Ratos Wistar , Vasodilatação/efeitos dos fármacos
8.
Mol Cell Biochem ; 355(1-2): 83-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21533769

RESUMO

Arginase constrains endothelial nitric oxide synthase activity by competing for the common substrate, L -Arginine. We have recently shown that inducible nitric oxide synthase (NOS2) S-nitrosates and activates arginase 1 (Arg1) leading to age-associated vascular dysfunction. Here, we demonstrate that a direct interaction of Arg1 with NOS2 is necessary for its S-nitrosation. The specific domain of NOS2 that mediates this interaction is identified. Disruption of this interaction in human aortic endothelial cells prevents Arg1 S-nitrosation and activation. Thus, disruption of NOS2-Arg1 interaction may represent a therapeutic strategy to attenuate age related vascular endothelial dysfunction.


Assuntos
Arginase/metabolismo , Nitratos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Envelhecimento/patologia , Animais , Aorta/citologia , Aorta/efeitos dos fármacos , Aorta/enzimologia , Linhagem Celular , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Ensaios Enzimáticos , Humanos , Imunoprecipitação , Interferon gama/farmacologia , Interferon gama/fisiologia , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/química , Óxido Nítrico Sintase Tipo II/genética , Nitrosação , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
9.
Atherosclerosis ; 214(2): 279-87, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21130456

RESUMO

AIMS: Arginase II regulates NOS activity by competing for the substrate l-arginine. Oxidized LDL (OxLDL) is a proatherogenic molecule that activates arginase II. We tested the hypotheses that OxLDL-dependent arginase II activation occurs through a specific receptor, and via a Rho GTPase effector mechanism that is inhibited by statins. METHODS AND RESULTS: Arginase II activation by OxLDL was attenuated following preincubation with the LOX-1 receptor-blocking antibody JTX92. This also prevented the dissociation of arginase II from microtubules. LOX-1(-/-) mice failed to exhibit the increased arginase II activity seen in WT mice fed a high cholesterol diet. Furthermore, endothelium from LOX-1(-/-) mice failed to demonstrate the diet-dependent reduction in NO and increase in ROS that were observed in WT mice. OxLDL induced Rho translocation to the membrane and Rho activation, and these effects were inhibited by pretreatment with JTX92 or statins. Transfection with siRNA for RhoA, or inhibition of ROCK both decreased OxLDL-stimulated arginase II activation. Preincubation with simvastatin or lovastatin blocked OxLDL-induced dissociation of arginase II from microtubules and prevented microtubule depolymerization. CONCLUSIONS: This study provides a new focus for preventive therapy for atherosclerotic disease by delineating a clearer path from OxLDL through the endothelial cell LOX-1 receptor, RhoA, and ROCK, to the activation of arginase II, downregulation of NO, and vascular dysfunction.


Assuntos
Arginase/metabolismo , Aterosclerose/enzimologia , Células Endoteliais/enzimologia , Lipoproteínas LDL/metabolismo , Receptores Depuradores Classe E/metabolismo , Transdução de Sinais , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Células Cultivadas , Colesterol na Dieta/metabolismo , Células Endoteliais/efeitos dos fármacos , Ativação Enzimática , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lovastatina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microtúbulos/metabolismo , Óxido Nítrico/metabolismo , Transporte Proteico , Interferência de RNA , Receptores Depuradores Classe E/deficiência , Receptores Depuradores Classe E/genética , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Superóxidos/metabolismo , Transfecção , Proteína rhoA de Ligação ao GTP/genética
10.
Nat Med ; 16(7): 767-73, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20473305

RESUMO

Rtp801 (also known as Redd1, and encoded by Ddit4), a stress-related protein triggered by adverse environmental conditions, inhibits mammalian target of rapamycin (mTOR) by stabilizing the TSC1-TSC2 inhibitory complex and enhances oxidative stress-dependent cell death. We postulated that Rtp801 acts as a potential amplifying switch in the development of cigarette smoke-induced lung injury, leading to emphysema. Rtp801 mRNA and protein were overexpressed in human emphysematous lungs and in lungs of mice exposed to cigarette smoke. The regulation of Rtp801 expression by cigarette smoke may rely on oxidative stress-dependent activation of the CCAAT response element in its promoter. We also found that Rtp801 was necessary and sufficient for nuclear factor-kappaB (NF-kappaB) activation in cultured cells and, when forcefully expressed in mouse lungs, it promoted NF-kappaB activation, alveolar inflammation, oxidative stress and apoptosis of alveolar septal cells. In contrast, Rtp801 knockout mice were markedly protected against acute cigarette smoke-induced lung injury, partly via increased mTOR signaling, and, when exposed chronically to cigarette smoke, against emphysema. Our data support the notion that Rtp801 may represent a major molecular sensor and mediator of cigarette smoke-induced lung injury.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Pulmão/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Enfisema Pulmonar/induzido quimicamente , Fumar/efeitos adversos , Fatores de Transcrição/fisiologia , Animais , Ativação Enzimática , Homeostase , Humanos , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Enfisema Pulmonar/genética , Serina-Treonina Quinases TOR , Fatores de Transcrição/genética
11.
J Appl Physiol (1985) ; 108(5): 1250-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20167676

RESUMO

Radiation exposure is associated with the development of various cardiovascular diseases. Although irradiation is known to cause elevated oxidant stress and chronic inflammation, both of which are detrimental to vascular function, the molecular mechanisms remain incompletely understood. We previously demonstrated that radiation causes endothelial dysfunction and increased vascular stiffness by xanthine oxidase (XO) activation. In this study, we investigated whether dietary inhibition of XO protects against radiation-induced vascular injury. We exposed 4-mo-old rats to a single dose of 0 or 5 Gy gamma radiation. These rats received normal drinking water or water containing 1 mM oxypurinol, an XO inhibitor. We measured XO activity and superoxide production in rat aorta and demonstrated that both were significantly elevated 2 wk after radiation exposure. However, oxypurinol treatment in irradiated rats prevented aortic XO activation and superoxide elevation. We next investigated endothelial function through fluorescent measurement of nitric oxide (NO) and vascular tension dose responses. Radiation reduced endothelium-dependent NO production in rat aorta. Similarly, endothelium-dependent vasorelaxation in the aorta of irradiated rats was significantly attenuated compared with the control group. Dietary XO inhibition maintained NO production at control levels and prevented the development of endothelial dysfunction. Furthermore, pulse wave velocity, a measure of vascular stiffness, increased by 1 day postirradiation and remained elevated 2 wk after irradiation, despite unchanged blood pressures. In oxypurinol-treated rats, pulse wave velocities remained unchanged from baseline throughout the experiment, signifying preserved vascular health. These findings demonstrate that XO inhibition can offer protection from radiation-induced endothelial dysfunction and cardiovascular complications.


Assuntos
Aorta/efeitos dos fármacos , Dieta , Endotélio Vascular/efeitos dos fármacos , Inibidores Enzimáticos/administração & dosagem , Oxipurinol/administração & dosagem , Lesões Experimentais por Radiação/prevenção & controle , Doenças Vasculares/prevenção & controle , Xantina Oxidase/antagonistas & inibidores , Animais , Aorta/enzimologia , Aorta/fisiopatologia , Aorta/efeitos da radiação , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Elasticidade , Endotélio Vascular/enzimologia , Endotélio Vascular/fisiopatologia , Endotélio Vascular/efeitos da radiação , Raios gama , Masculino , Óxido Nítrico/metabolismo , Fluxo Pulsátil , Lesões Experimentais por Radiação/enzimologia , Lesões Experimentais por Radiação/fisiopatologia , Ratos , Ratos Sprague-Dawley , Fluxo Sanguíneo Regional , Superóxidos/metabolismo , Fatores de Tempo , Ultrassonografia Doppler , Doenças Vasculares/enzimologia , Doenças Vasculares/fisiopatologia , Vasodilatação/efeitos dos fármacos , Vasodilatação/efeitos da radiação , Vasodilatadores/farmacologia , Irradiação Corporal Total , Xantina Oxidase/metabolismo
12.
J Mol Med (Berl) ; 85(12): 1317-24, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18040654

RESUMO

The lung is both the conduit for oxygen uptake and is also affected by hypoxia and hypoxia signaling. Decreased ventilatory drive, airway obstructive processes, intra-alveolar exudates, septal thickening by edema, inflammation, fibrosis, or damage to alveolar capillaries will all interpose a significant and potentially life-threatening barrier to proper oxygenation, therefore enhancing the alveolar/arterial pO2 gradient. These processes result in decreased blood and tissue oxygenation. This review addresses the relationship of hypoxia with lung development and with lung diseases. We particularly focus on molecular mechanisms underlying hypoxia-driven physiological and pathophysiological lung processes, specifically in the infant lung, pulmonary hypertension, and chronic obstructive pulmonary disease.


Assuntos
Hipóxia/metabolismo , Pneumopatias/metabolismo , Pulmão/metabolismo , Animais , Doença Crônica , Humanos , Hipertensão Pulmonar/metabolismo , Hipóxia/patologia , Hipóxia/fisiopatologia , Fator 1 Induzível por Hipóxia/metabolismo , Recém-Nascido , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Pulmão/fisiopatologia , Pneumopatias/patologia , Pneumopatias/fisiopatologia , Oxigênio/metabolismo , Circulação Pulmonar , Doença Pulmonar Obstrutiva Crônica/metabolismo , Troca Gasosa Pulmonar , Síndrome do Desconforto Respiratório do Recém-Nascido/metabolismo , Transdução de Sinais
13.
J Am Soc Nephrol ; 17(3): 637-47, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16452497

RESUMO

Polycystin-1 (PC-1), the PKD1 gene product, is a large receptor whose expression in renal epithelial cells results in resistance to apoptosis and tubulogenesis, a model consistent with the phenotype observed in patients. This study links PC-1 expression to a signaling pathway that is known to be both antiapoptotic and important for normal tubulogenesis. This study found that PC-1 expression results in phosphorylation of Akt and downstream effectors and that phosphatidylinositol 3-kinase (PI3-K) inhibitors prevent this process. In addition, it is shown that dominant negative Akt can revert PC-1-induced protection from apoptosis. Furthermore, it was observed that increased PI3-K beta activity in PC-1-expressing MDCK cells seems to be dependent on both tyrosine-kinase activity and heterotrimeric G proteins. It also was found that PC-1-induced tubulogenesis is inhibited by PI3-K inhibitors. Taken together, these data suggest that the PI3-K/Akt cascade may be a central modulator of PC-1 function and that its deregulation might be important in autosomal dominant polycystic kidney disease.


Assuntos
Apoptose/fisiologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Canais de Cátion TRPP/metabolismo , Proliferação de Células , Células Cultivadas , Mesângio Glomerular/citologia , Mesângio Glomerular/metabolismo , Humanos , Sensibilidade e Especificidade , Transdução de Sinais
14.
Glycoconj J ; 20(5): 331-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15229397

RESUMO

Although low concentrations (10 microg/ml) of oxidized LDL density lipoproteins (Ox-LDL) and minimally modified LDL (MM-LDL) can stimulate the proliferation of aortic smooth muscle cells the biologically active component responsible for this phenomena has not been identified. Here we report that the 1-palmitoyl-2-(5-oxovaleroyl)-sn-glycero-4-phosphocholine (m/e594.3) (POVPC) present in MM-LDL but not 1-palmitoyl-2-glutaryl-sn-glycero-3-phophochline (m/e610.2)(PGPC) can stimulate the activity of UDP-galactose:glucosylceramide (beta 1-->4) galactosyltransferase (GalT-2) and produce lactosyceramide (LacCer). LacCer, in turn, generated superoxide radicals (O(2)(.-)). This is accompanied by the phosphorylation/activation of a cytosolic transcriptional factor p(44) MAPK and the subsequent proliferation of human aortic smooth muscle cells. D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (D-PDMP), an inhibitor of GalT-2, impaired the induction of GalT-2 activity, O(2)(.-)generation, and cell proliferation. Thus POVPC may serve as a surrogate in MM-LDL mediated induction of aortic smooth muscle cells (A-SMC) proliferation via GalT-2 activation. The LacCer produced as a consequence of GalT-2 activation may serve as a lipid second messenger in the activation of an oxidant sensitive transcriptional pahtway that ultimately leads to cell proliferation and may contribute to the pathophysiology of atherosclerosis.


Assuntos
Aorta/citologia , Lipoproteínas LDL/química , Lipoproteínas LDL/farmacologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Galactosiltransferases/antagonistas & inibidores , Galactosiltransferases/metabolismo , Humanos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Oxirredução , Éteres Fosfolipídicos/farmacologia , Fosfolipídeos/farmacologia , Proteínas de Plantas , Esfingolipídeos/farmacologia , Superóxidos/metabolismo
15.
Proc Natl Acad Sci U S A ; 99(26): 16981-6, 2002 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-12482949

RESUMO

Polycystin-1 plays an essential role in renal tubular morphogenesis, and disruption of its function causes cystogenesis in human autosomal-dominant polycystic kidney disease (ADPKD). We demonstrated that polycystin-1 undergoes cleavage at G protein coupled receptor proteolytic site in a process that requires the receptor for egg jelly domain. Most of the N-terminal fragment remains tethered at the cell surface, although a small amount is secreted. PKD1-associated mutations in the receptor for egg jelly domain disrupt cleavage, abolish the ability of polycystin-1 to activate signal transducer and activator of transcription-1, and induce tubulogenesis in vitro. We conclude that the cleavage of polycystin-1 is likely essential for its biologic activity.


Assuntos
Mutação , Rim Policístico Autossômico Dominante/genética , Proteínas/metabolismo , Receptores de Superfície Celular/fisiologia , Sequência de Aminoácidos , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas/química , Receptores de Superfície Celular/química , Transdução de Sinais , Canais de Cátion TRPP
16.
Cell ; 109(2): 157-68, 2002 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-12007403

RESUMO

Autosomal dominant polycystic kidney disease is characterized by cyst formation in the kidney and other organs and results from mutations of PKD1 or PKD2. Previous studies suggest that their gene products have an important role in growth regulation. We now show that expression of polycystin-1 activates the JAK-STAT pathway, thereby upregulating p21(waf1) and inducing cell cycle arrest in G0/G1. This process requires polycystin-2, a channel protein, as an essential cofactor. Mutations that disrupt polycystin-1/2 binding prevent activation of the pathway. Mouse embryos lacking Pkd1 have defective STAT1 phosphorylation and p21(waf1) induction. These results suggest that one function of the polycystin-1/2 complex is to regulate the JAK/STAT pathway and explain how mutations of either gene can result in dysregulated growth.


Assuntos
Ciclo Celular/genética , Ciclinas/genética , Rim Policístico Autossômico Dominante/genética , Proteínas Tirosina Quinases/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas , Animais , Divisão Celular/genética , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/genética , Feminino , Fase G1/genética , Humanos , Janus Quinase 2 , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Mutação/genética , Rim Policístico Autossômico Dominante/metabolismo , Proteínas/genética , Fase de Repouso do Ciclo Celular/genética , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transdução de Sinais/genética , Canais de Cátion TRPP , Transativadores/genética
17.
J Biol Chem ; 277(19): 16396-402, 2002 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-11861654

RESUMO

Sialic acid containing glycosphingolipids (gangliosides) are expressed on the surface of all mammalian cells and have been implicated in regulating various biological phenomena; however, the detailed signaling mechanisms involved in this process are not known. We report here a novel aspect of disialoganglioside, GD3-mediated regulation of cell proliferation and cell death via the recruitment of reactive oxygen species (ROS). A low concentration (2.5-10 microm) of GD3, incubated with human aortic smooth muscle cells for a short period of time (10-30 min), stimulates superoxide generation via the activation of both NADPH oxidase and NADH oxidase activity. This leads to downstream signaling leading to cell proliferation and apoptosis. However, [(3)H]GD3 incubated with the cells under such conditions was found in a trypsin-sensitive fraction that was separable from endogenous GD3. The exact mechanism causing ROS generation and downstream signaling remains to be elucidated. The uptake of GD3 was accompanied by a 2.5-fold stimulation in the activity of mitogen-activated protein (MAP) kinase and 5-fold stimulation in cell proliferation. Preincubation of cells with membrane-permeable antioxidants, pyrrolidine dithiocarbamate, and N-acetylcysteine abrogated the superoxide generation and cell proliferation. In contrast, at higher concentrations (50-200 microm) GD3 inhibited the generation of superoxides but markedly stimulated the generation of nitric oxide (NO) (10-fold compared with control). This in turn stimulated mitochondrial cytochrome c release and intrachromosomal DNA fragmentation, which lead to apoptosis. In sum, at a low concentration, GD3 recruits superoxides to activate p44 MAPK and stimulates cell proliferation. In contrast, at high concentrations GD3 recruits nitric oxide to scavenge superoxide radicals that triggered signaling events that led to apoptosis. These observations might have relevance in regard to the potential role of GD3 in aortic smooth muscle cell proliferation and apoptosis that may contribute to plaque rupture in atherosclerosis.


Assuntos
Aorta/citologia , Apoptose , Endotélio Vascular/citologia , Gangliosídeos/metabolismo , Gangliosídeos/fisiologia , Músculo Liso/citologia , Espécies Reativas de Oxigênio , Arteriosclerose , Divisão Celular , Células Cultivadas , Grupo dos Citocromos c/metabolismo , Fragmentação do DNA , Relação Dose-Resposta a Droga , Humanos , Cinética , Sistema de Sinalização das MAP Quinases , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Complexos Multienzimáticos/metabolismo , NADH NADPH Oxirredutases/metabolismo , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Testes de Precipitina , Transdução de Sinais , Frações Subcelulares/metabolismo , Superóxidos/metabolismo , Fatores de Tempo , Tripsina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA