Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Immunol ; 191(5): 2290-8, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23913966

RESUMO

Arginase I is a marker of murine M2 macrophages and is highly expressed in many inflammatory diseases. The basis for high arginase I expression in macrophages in vivo is incompletely understood but likely reflects integrated responses to combinations of stimuli. Our objective was to elucidate mechanisms involved in modulating arginase I induction by IL-4, the prototypical activator of M2 macrophages. IL-4 and 8-bromo-cAMP individually induce arginase I, but together they rapidly and synergistically induce arginase I mRNA, protein, and promoter activity in murine macrophage cells. Arginase I induction by IL-4 requires binding of the transcription factors STAT6 and C/EBPß to the IL-4 response element of the arginase I gene. Chromatin immunoprecipitation showed that the synergistic response involves binding of both transcription factors to the IL-4 response element at levels significantly greater than in response to IL-4 alone. The results suggest that C/EBPß is a limiting factor for the level of STAT6 bound to the IL-4 response element. The enhanced binding in the synergistic response was not due to increased expression of either STAT6 or C/EBPß but was correlated primarily with increased nuclear abundance of C/EBPß. Our findings also suggest that induction of arginase I expression is stochastic; that is, differences in induction reflect differences in probability of transcriptional activation and not simply differences in rate of transcription. Results of the present study also may be useful for understanding mechanisms underlying regulated expression of other genes in macrophages and other myeloid-derived cells in health and disease.


Assuntos
Arginase/biossíntese , AMP Cíclico/metabolismo , Regulação da Expressão Gênica/imunologia , Interleucina-4/metabolismo , Ativação de Macrófagos/fisiologia , Macrófagos/metabolismo , Regiões Promotoras Genéticas , Animais , Arginase/genética , Arginase/imunologia , Linhagem Celular , Imunoprecipitação da Cromatina , AMP Cíclico/imunologia , Immunoblotting , Interleucina-4/imunologia , Macrófagos/imunologia , Camundongos , Fenótipo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Transfecção
2.
J Immunol ; 191(2): 773-84, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23749634

RESUMO

Macrophages in granulomas are both antimycobacterial effector and host cell for Mycobacterium tuberculosis, yet basic aspects of macrophage diversity and function within the complex structures of granulomas remain poorly understood. To address this, we examined myeloid cell phenotypes and expression of enzymes correlated with host defense in macaque and human granulomas. Macaque granulomas had upregulated inducible and endothelial NO synthase (iNOS and eNOS) and arginase (Arg1 and Arg2) expression and enzyme activity compared with nongranulomatous tissue. Immunohistochemical analysis indicated macrophages adjacent to uninvolved normal tissue were more likely to express CD163, whereas epithelioid macrophages in regions where bacteria reside strongly expressed CD11c, CD68, and HAM56. Calprotectin-positive neutrophils were abundant in regions adjacent to caseum. iNOS, eNOS, Arg1, and Arg2 proteins were identified in macrophages and localized similarly in granulomas across species, with greater eNOS expression and ratio of iNOS/Arg1 expression in epithelioid macrophages as compared with cells in the lymphocyte cuff. iNOS, Arg1, and Arg2 expression in neutrophils was also identified. The combination of phenotypic and functional markers support that macrophages with anti-inflammatory phenotypes localized to outer regions of granulomas, whereas the inner regions were more likely to contain macrophages with proinflammatory, presumably bactericidal, phenotypes. Together, these data support the concept that granulomas have organized microenvironments that balance antimicrobial anti-inflammatory responses to limit pathology in the lungs.


Assuntos
Arginase/metabolismo , Granuloma/imunologia , Macrófagos/citologia , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Tuberculose/imunologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Antígeno CD11c/metabolismo , Microambiente Celular , Humanos , Complexo Antígeno L1 Leucocitário/metabolismo , Pulmão/microbiologia , Pulmão/patologia , Macaca , Mycobacterium tuberculosis/imunologia , Células Mieloides , Neutrófilos/metabolismo , Isoformas de Proteínas/metabolismo , Receptores de Superfície Celular/metabolismo , Tuberculose/microbiologia , Tuberculose/patologia
3.
FASEB J ; 26(1): 376-86, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21926236

RESUMO

Adenosine has been implicated in suppressing the proinflammatory responses of classically activated macrophages induced by Th1 cytokines. Alternative macrophage activation is induced by the Th2 cytokines interleukin (IL)-4 and IL-13; however, the role of adenosine in governing alternative macrophage activation is unknown. We show here that adenosine treatment of IL-4- or IL-13-activated macrophages augments the expression of alternative macrophage markers arginase-1, tissue inhibitor of matrix metalloproteinase-1 (TIMP-1), and macrophage galactose-type C-type lectin-1. The stimulatory effect of adenosine required primarily A(2B) receptors because the nonselective adenosine receptor agonist 5'-N-ethylcarboxamidoadenosine (NECA) increased both arginase activity (EC(50)=261.8 nM) and TIMP-1 production (EC(50)=80.67 nM), and both pharmacologic and genetic blockade of A(2B) receptors prevented the effect of NECA. A(2A) receptors also contributed to the adenosine augmentation of IL-4-induced TIMP-1 release, as both adenosine and NECA were less efficacious in augmenting TIMP-1 release by A(2A) receptor-deficient than control macrophages. Of the transcription factors known to drive alternative macrophage activation, CCAAT-enhancer-binding protein ß was required, while cAMP response element-binding protein and signal transducer and activator of transcription 6 were dispensable in mediating the effect of adenosine. We propose that adenosine receptor activation suppresses inflammation and promotes tissue restitution, in part, by promoting alternative macrophage activation.


Assuntos
Adenosina/metabolismo , Inflamação/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptor A2B de Adenosina/metabolismo , Adenosina/farmacologia , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Animais , Arginase/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Espaço Extracelular/metabolismo , Inflamação/imunologia , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/imunologia , Receptor A2B de Adenosina/genética , Receptor A2B de Adenosina/imunologia , Fator de Transcrição STAT6/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Vasodilatadores/farmacologia
4.
Circ Res ; 109(5): 492-501, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21757649

RESUMO

RATIONALE: Activation of liver X receptors (LXRs) inhibits the progression of atherosclerosis and promotes regression of existing lesions. In addition, LXRα levels are high in regressive plaques. Macrophage arginase 1 (Arg1) expression is inversely correlated with atherosclerosis progression and is markedly decreased in foam cells within the lesion. OBJECTIVE: To investigate LXRα regulation of Arg1 expression in cultured macrophages and atherosclerotic regressive lesions. METHODS AND RESULTS: We found that Arg1 expression is enhanced in CD68+ cells from regressive versus progressive lesions in a murine aortic arch transplant model. In cultured macrophages, ligand-activated LXRα markedly enhances basal and interleukin-4-induced Arg1 mRNA and protein expression as well as promoter activity. This LXRα-enhanced Arg1 expression correlates with a reduction in nitric oxide levels. Moreover, Arg1 expression within regressive atherosclerotic plaques is LXRα-dependent, as enhanced expression of Arg1 in regressive lesions is impaired in LXRα-deficient CD68+ cells. LXRα does not bind to the Arg1 promoter but instead promotes the interaction between PU.1 and interferon regulatory factor (IRF)8 transcription factors and induces their binding of a novel composite element. Accordingly, knockdown of either IRF8 or PU.1 strongly impairs LXRα regulation of Arg1 expression in macrophage cells. Finally, we demonstrate that LXRα binds the IRF8 locus and its activation increases IRF8 mRNA and protein levels in these cells. CONCLUSIONS: This work implicates Arg1 in atherosclerosis regression and identifies LXRα as a novel regulator of Arg1 and IRF8 in macrophages. Furthermore, it provides a unique molecular mechanism by which LXRα regulates macrophage target gene expression through PU.1 and IRF8.


Assuntos
Arginase/metabolismo , Fatores Reguladores de Interferon/fisiologia , Macrófagos/metabolismo , Receptores Nucleares Órfãos/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Animais , Arginase/biossíntese , Arginase/genética , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Linhagem Celular , Marcação de Genes/métodos , Loci Gênicos , Fatores Reguladores de Interferon/antagonistas & inibidores , Fatores Reguladores de Interferon/metabolismo , Receptores X do Fígado , Macrófagos/enzimologia , Camundongos , Camundongos Knockout , Receptores Nucleares Órfãos/deficiência , Receptores Nucleares Órfãos/genética , Ligação Proteica/genética , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Transativadores/antagonistas & inibidores , Transativadores/metabolismo
5.
Methods Enzymol ; 440: 221-30, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18423220

RESUMO

Of all arginine catabolic enzymes, the arginases and nitric oxide (NO) synthases are the ones that are of greatest interest to many investigators. Mammalian arginases catalyze the hydrolysis of arginine to ornithine and urea and are composed of two distinct isozymes: arginase I, located within the cytosol, and arginase II, located within mitochondria. The arginases not only can inhibit NO synthesis by reducing arginine availability, but also can promote the synthesis of polyamines or proline via production of the common precursor ornithine. Because of their inducibility in many cell types and to their potential impact on multiple biochemical pathways in health and disease, there is growing interest in assays of arginase activity. Although arginase activity may be determined by either spectrophotometric or radiochemical assays, radiochemical assays afford greater sensitivity and do not require correction for any ornithine or urea that may be present in the samples. Part of the arginase assay protocol described in this chapter also can be used for radiochemical assays of enzymes that catalyze decarboxylation reactions. No activity assay currently available is capable of distinguishing the arginase isozymes.


Assuntos
Arginase/análise , Arginina/metabolismo , Mamíferos/metabolismo , Animais , Arginase/fisiologia , Ativação Enzimática/fisiologia , Humanos , Especificidade por Substrato/fisiologia
6.
J Toxicol Environ Health A ; 70(2): 118-27, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17365572

RESUMO

Arginase induction was reported in several inflammatory lung diseases, suggesting that this may be a common feature underlying the pathophysiology of such diseases. As little is known regarding arginase expression in silicosis, the induction and cellular localization of arginase were elucidated in lungs of Sprague-Dawley rats 24 h following exposure to varying doses of silica by intratracheal instillation. Arginase expression was evaluated by activity assay, quantification of arginase I and arginase II mRNA levels using real-time polymerase chain reaction (PCR), and immunohistochemistry. Analyses of cells and fluid obtained by bronchoalveolar lavage (BAL) showed that markers of pulmonary inflammation, tissue damage, activation of alveolar macrophages (AM) and NO production were significantly increased by all silica doses. Arginase activity was increased also in AMs isolated from BAL fluid of silica-treated rats. Silica produced two- and three-fold increases in arginase activity of whole lung at doses of 1 and 5 mg/100 g body weight, respectively. Levels of arginase I mRNA, but not of arginase II mRNA, were similarly elevated. In control lungs, arginase I immunoreactivity was observed only in AMs sparsely dispersed throughout the lung; no inducible nitric oxide synthase (iNOS) immunoreactivity was detected. In silica-treated lungs, arginase I and iNOS were co-expressed in most AMs that were abundantly clustered at inflammatory foci. The rapid induction of arginase I expression in inflammatory lung cells, similar to induction of arginase in other inflammatory lung diseases, implicates elevated arginase activity as a factor in the development of lung damage following exposure to silica.


Assuntos
Arginase/metabolismo , Pulmão/efeitos dos fármacos , Dióxido de Silício/toxicidade , Albuminas/metabolismo , Animais , Arginase/genética , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Desidrogenase/metabolismo , Contagem de Leucócitos , Pulmão/metabolismo , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Masculino , Neutrófilos/efeitos dos fármacos , Neutrófilos/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
7.
Gene ; 353(1): 98-106, 2005 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-15922518

RESUMO

Arginine metabolism in macrophages during infection and inflammation is complex, owing to differential regulation of inducible nitric oxide synthase (iNOS) and arginases by cytokines and other agents. Changes in levels of Th2 cytokines such as interleukin-4 (IL-4) can play important roles in these conditions via effects on arginine metabolism. IL-4 alters macrophage arginine metabolism by inducing arginase I expression and inhibiting nitric oxide production. To determine the molecular basis for induction of arginase I, the promoter of the murine arginase I gene was cloned and analyzed by transfection in RAW 264.7 macrophage cells. IL-4 induction required a composite response element containing STAT6 and C/EBP sites located 2.86 kb upstream of the transcription start site. Competition experiments showed that STAT6 and C/EBPbeta bind to the STAT6 and C/EBP sites non-cooperatively. Elucidation of the mechanisms involved in regulation of arginase I transcription may provide a basis for developing strategies to modulate arginase expression in Th2 cytokine-predominant diseases.


Assuntos
Arginase/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Interleucina-4/farmacologia , Elementos de Resposta/genética , Transativadores/metabolismo , Animais , Sequência de Bases , Sítios de Ligação/genética , Ligação Competitiva , Linhagem Celular , DNA/genética , DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Luciferases/genética , Luciferases/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT6 , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica/efeitos dos fármacos , Transfecção
8.
PLoS One ; 7(7): e39487, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22829869

RESUMO

BACKGROUND: Cardiovascular disorders associated with endothelial dysfunction, such as atherosclerosis, have decreased nitric oxide (NO) bioavailability. Arginase in the vasculature can compete with eNOS for L-arginine and has been implicated in atherosclerosis. The aim of this study was to evaluate the effect of endothelial-specific elevation of arginase II expression on endothelial function and the development of atherosclerosis. METHODOLOGY/PRINCIPAL FINDINGS: Transgenic mice on a C57BL/6 background with endothelial-specific overexpression of human arginase II (hArgII) gene under the control of the Tie2 promoter were produced. The hArgII mice had elevated tissue arginase activity except in liver and in resident peritoneal macrophages, confirming endothelial specificity of the transgene. Using small-vessel myography, aorta from these mice exhibited endothelial dysfunction when compared to their non-transgenic littermate controls. The blood pressure of the hArgII mice was 17% higher than their littermate controls and, when crossed with apoE -/- mice, hArgII mice had increased aortic atherosclerotic lesions. CONCLUSION: We conclude that overexpression of arginase II in the endothelium is detrimental to the cardiovascular system.


Assuntos
Arginase/metabolismo , Aterosclerose/enzimologia , Aterosclerose/patologia , Endotélio Vascular/enzimologia , Hipertensão/enzimologia , Animais , Arginase/genética , Aterosclerose/genética , Pressão Sanguínea/fisiologia , Western Blotting , Endotélio Vascular/patologia , Hipertensão/genética , Hipertensão/patologia , Macrófagos Peritoneais , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Diabetes ; 60(11): 3015-22, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21926276

RESUMO

OBJECTIVE: To determine 1) whether renal arginase activity or expression is increased in diabetes and 2) whether arginase plays a role in development of diabetic nephropathy (DN). RESEARCH DESIGN AND METHODS: The impact of arginase activity and expression on renal damage was evaluated in spontaneously diabetic Ins2(Akita) mice and in streptozotocin (STZ)-induced diabetic Dilute Brown Agouti (DBA) and arginase-2-deficient mice (Arg2(-/-)). RESULTS: Pharmacological blockade or genetic deficiency of arginase-2 conferred kidney protection in Ins2(Akita) mice or STZ-induced diabetic renal injury. Blocking arginases using S-(2-boronoethyl)-L-cysteine for 9 weeks in Ins2(Akita) mice or 6 weeks in STZ-induced diabetic DBA mice significantly attenuated albuminuria, the increase in blood urea nitrogen, histopathological changes, and kidney macrophage recruitment compared with vehicle-treated Ins2(Akita) mice. Furthermore, kidney arginase-2 expression increased in Ins2(Akita) mice compared with control. In contrast, arginase-1 expression was undetectable in kidneys under normal or diabetes conditions. Arg2(-/-) mice mimicked arginase blockade by reducing albuminuria after 6 and 18 weeks of STZ-induced diabetes. In wild-type mice, kidney arginase activity increased significantly after 6 and 18 weeks of STZ-induced diabetes but remained very low in STZ-diabetic Arg2(-/-) mice. The increase in kidney arginase activity was associated with a reduction in renal medullary blood flow in wild-type mice after 6 weeks of STZ-induced diabetes, an effect significantly attenuated in diabetic Arg2(-/-) mice. CONCLUSIONS: These findings indicate that arginase-2 plays a major role in induction of diabetic renal injury and that blocking arginase-2 activity or expression could be a novel therapeutic approach for treatment of DN.


Assuntos
Arginase/metabolismo , Nefropatias Diabéticas/metabolismo , Rim/metabolismo , Albuminúria/etiologia , Albuminúria/prevenção & controle , Animais , Arginase/antagonistas & inibidores , Arginase/genética , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/complicações , Nefropatias Diabéticas/patologia , Nefropatias Diabéticas/fisiopatologia , Nefropatias Diabéticas/prevenção & controle , Modelos Animais de Doenças , Inibidores Enzimáticos/toxicidade , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Histiócitos/efeitos dos fármacos , Histiócitos/patologia , Hiperglicemia/etiologia , Rim/irrigação sanguínea , Rim/patologia , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Mutantes , Camundongos Transgênicos , Terapia de Alvo Molecular , RNA Mensageiro/metabolismo , Fluxo Sanguíneo Regional , Estreptozocina/toxicidade
10.
PLoS One ; 5(12): e15253, 2010 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-21151916

RESUMO

Increased catabolism of arginine by arginase is increasingly viewed as an important pathophysiological factor in cardiovascular disease, including atherosclerosis induced by high cholesterol diets. Whereas previous studies have focused primarily on effects of high cholesterol diets on arginase expression and arginine metabolism in specific blood vessels, there is no information regarding the impact of lipid diets on arginase activity or arginine bioavailability at a systemic level. We, therefore, evaluated the effects of high fat (HF) and high fat-high cholesterol (HC) diets on arginase activity in plasma and tissues and on global arginine bioavailability (defined as the ratio of plasma arginine to ornithine + citrulline) in apoE(-/-) and wild-type C57BL/6J mice. HC and HF diets led to reduced global arginine bioavailability in both strains. The HC diet resulted in significantly elevated plasma arginase in both strains, but the HF diet increased plasma arginase only in apoE(-/-) mice. Elevated plasma arginase activity correlated closely with increased alanine aminotransferase levels, indicating that liver damage was primarily responsible for elevated plasma arginase. The HC diet, which promotes atherogenesis, also resulted in increased arginase activity and expression of the type II isozyme of arginase in multiple tissues of apoE(-/-) mice only. These results raise the possibility that systemic changes in arginase activity and global arginine bioavailability may be contributing factors in the initiation and/or progression of cardiovascular disease.


Assuntos
Apolipoproteínas E/genética , Arginase/metabolismo , Arginina/farmacologia , Colesterol/metabolismo , Alanina Transaminase/metabolismo , Ração Animal , Animais , Arginase/sangue , Disponibilidade Biológica , Citrulina/genética , Dieta , Isoenzimas/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ornitina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Am J Physiol Lung Cell Mol Physiol ; 290(3): L534-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16257997

RESUMO

Arginase is greatly elevated in asthma and is thought to play a role in the pathophysiology of this disease. As inhibitors of phosphodiesterase 4 (PDE4), the predominant PDE in macrophages, elevate cAMP levels and reduce inflammation, they have been proposed for use in treatment of asthma and chronic obstructive pulmonary disease. As cAMP is an inducer of arginase, we tested the hypothesis that a PDE4 inhibitor would enhance macrophage arginase induction by key cytokines implicated in asthma and other pulmonary diseases. RAW 264.7 cells were stimulated with IL-4 or transforming growth factor (TGF)-beta, with and without the PDE4 inhibitor rolipram. IL-4 and TGF-beta increased arginase activity 16- and 5-fold, respectively. Rolipram alone had no effect but when combined with IL-4 and TGF-beta synergistically enhanced arginase activity by an additional 15- and 5-fold, respectively. The increases in arginase I protein and mRNA levels mirrored increases in arginase activity. Induction of arginase II mRNA was also enhanced by rolipram but to a much lesser extent than arginase I. Unlike its effect in RAW 264.7 cells, IL-4 alone did not increase arginase activity in human alveolar macrophages (AM) from healthy volunteers. However, combining IL-4 with agents to induce cAMP levels induced arginase activity in human AM significantly above the level obtained with cAMP-inducing agents alone. In conclusion, agents that elevate cAMP significantly enhance induction of arginase by cytokines. Therefore, consequences of increased arginase expression should be evaluated whenever PDE inhibitors are proposed for treatment of inflammatory disorders in which IL-4 and/or TGF-beta predominate.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , Arginase/biossíntese , Interleucina-4/farmacologia , Macrófagos Alveolares/enzimologia , Fator de Crescimento Transformador beta/farmacologia , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Animais , Antineoplásicos/farmacologia , Arginase/genética , Líquido da Lavagem Broncoalveolar/química , Linhagem Celular , AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Sinergismo Farmacológico , Indução Enzimática , Humanos , Camundongos , Inibidores de Fosfodiesterase/farmacologia , Regiões Promotoras Genéticas/genética , Rolipram/farmacologia
12.
Biochem Biophys Res Commun ; 294(4): 749-52, 2002 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-12061769

RESUMO

The urea cycle, which involves enzymes located in both the mitochondrion and cytoplasm, requires transport of ornithine and citrulline across the mitochondrial membrane by the ornithine/citrulline antiporter ORNT1. Expression of the urea cycle enzymes can change dramatically in response to hormones, but it is not known whether ORNT1 expression also is hormonally regulated. This study therefore tested the hypothesis that ORNT1 mRNA levels in hepatocytes are induced by cAMP and glucocorticoid as are the urea cycle enzyme mRNAs. ORNT1 mRNA was rapidly induced by a cAMP analog and dexamethasone in cultured rat hepatocytes and there was a strong synergistic response to a combination of these agents. Ongoing protein synthesis was required for induction of ORNT1 mRNA by dexamethasone but not by cAMP, suggesting that the dexamethasone response required an accessory factor. Thus, hormonal regulation of ORNT1 mRNA in hepatocytes is coordinated with that of mRNAs encoding the urea cycle enzymes.


Assuntos
Dexametasona/farmacologia , Glucocorticoides/farmacologia , Mitocôndrias Hepáticas/metabolismo , Biossíntese de Proteínas , Ativação Transcricional , Sistemas de Transporte de Aminoácidos Básicos , Animais , Transporte Biológico , Células Cultivadas , Citrulina/metabolismo , AMP Cíclico/metabolismo , Cicloeximida/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Masculino , Proteínas de Transporte da Membrana Mitocondrial , Modelos Biológicos , Ornitina/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Proteínas/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Ureia/metabolismo
13.
Am J Respir Crit Care Med ; 168(1): 63-9, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12626350

RESUMO

Pulmonary hypertension is a life-threatening complication of sickle cell disease. L-Arginine is the nitrogen donor for synthesis of nitric oxide, a potent vasodilator that is deficient during times of sickle cell crisis. This deficiency may play a role in pulmonary hypertension. The enzyme arginase hydrolyzes arginine to ornithine and urea, and thus, it may compete with nitric oxide synthase, leading to decreased nitric oxide production. Nitric oxide therapy by inhalation has improved pulmonary hypertension associated with acute chest syndrome in sickle cell disease, and several studies demonstrate therapeutic benefits of arginine therapy for primary and secondary pulmonary hypertension. We sought to determine the effects of arginine therapy on pulmonary hypertension in patients with sickle cell disease. Arginase activity was also determined. Oral arginine produced a 15.2% mean reduction in estimated pulmonary artery systolic pressure (63.9 +/- 13 to 54.2 +/- 12 mm Hg, p = 0.002) after 5 days of therapy in 10 patients. Arginase activity was elevated almost twofold (p = 0.07) in patients with pulmonary hypertension and may limit arginine bioavailability. With limited treatment options and a high mortality rate for patients with sickle cell disease who develop pulmonary hypertension, arginine is a promising new therapy that warrants further investigation.


Assuntos
Anemia Falciforme/complicações , Arginina/uso terapêutico , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Administração Oral , Adolescente , Adulto , Aminoácidos/sangue , Arginase/sangue , Arginase/efeitos dos fármacos , Arginina/metabolismo , Disponibilidade Biológica , Estudos de Casos e Controles , Ecocardiografia , Feminino , Humanos , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Masculino , Pessoa de Meia-Idade , Óxido Nítrico/metabolismo , Ornitina/sangue , Oximetria , Pressão Propulsora Pulmonar/efeitos dos fármacos , Resultado do Tratamento
14.
J Pediatr Hematol Oncol ; 25(8): 629-34, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12902916

RESUMO

PURPOSE: Recent data suggest that hydroxyurea (HU) increases the production of nitric oxide (NO), a potent vasodilator. NO is normally metabolized from l-Arginine (Arg). However, in vitro and animal experiments suggest that HU is the NO donor itself. In contrast, a recent study indicates that nitric oxide synthase (NOS) may play a role. Since adults with sickle cell disease (SCD) are Arg-deficient, Arg availability may limit the ability of HU to maximally impact NO production if an NOS mechanism is involved. The authors have previously shown that Arg supplementation alone induces a paradoxical decrease in NO metabolite (NO(x)) production. METHODS: The authors studied the effects of HU and Arg supplementation on NO(x) production. HU alone or HU + Arg was administered to patients with SCD at steady state, and sequential levels of Arg, serum NO(x) and exhaled NO were followed over 4 hours. RESULTS: After HU + Arg, all patients demonstrated a significant increase in serum NO(x) production within 2 hours. When the same patients were treated with HU alone (5.1 +/- 2 micromol/L), a mixed response occurred. NO(x) levels increased in four patients and decreased in one patient (-23.3 micromol/L). CONCLUSIONS: While Arg alone does not increase serum NO(x) production in SCD patients at steady state, it does when given together with HU. Hence, co-administration of Arg with HU may augment the NO(x) response in SCD and improve utilization of Arg in patients at steady state.


Assuntos
Anemia Falciforme/tratamento farmacológico , Antineoplásicos/farmacologia , Arginina/farmacologia , Sequestradores de Radicais Livres/análise , Hidroxiureia/farmacologia , Óxido Nítrico/análise , Adolescente , Adulto , Anemia Falciforme/complicações , Antineoplásicos/administração & dosagem , Arginina/administração & dosagem , Arginina/metabolismo , Criança , Interações Medicamentosas , Quimioterapia Combinada , Feminino , Humanos , Hidroxiureia/administração & dosagem , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA