Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 14(10): 686, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37852977

RESUMO

Ineffective hematopoiesis is a hallmark of myelodysplastic syndromes (MDS). Hematopoietic alterations in MDS patients strictly correlate with microenvironment dysfunctions, eventually affecting also the mesenchymal stromal cell (MSC) compartment. Stromal cells are indeed epigenetically reprogrammed to cooperate with leukemic cells and propagate the disease as "tumor unit"; therefore, changes in MSC epigenetic profile might contribute to the hematopoietic perturbations typical of MDS. Here, we unveil that the histone variant macroH2A1 (mH2A1) regulates the crosstalk between epigenetics and inflammation in MDS-MSCs, potentially affecting their hematopoietic support ability. We show that the mH2A1 splicing isoform mH2A1.1 accumulates in MDS-MSCs, correlating with the expression of the Toll-like receptor 4 (TLR4), an important pro-tumor activator of MSC phenotype associated to a pro-inflammatory behavior. MH2A1.1-TLR4 axis was further investigated in HS-5 stromal cells after ectopic mH2A1.1 overexpression (mH2A1.1-OE). Proteomic data confirmed the activation of a pro-inflammatory signature associated to TLR4 and nuclear factor kappa B (NFkB) activation. Moreover, mH2A1.1-OE proteomic profile identified several upregulated proteins associated to DNA and histones hypermethylation, including S-adenosylhomocysteine hydrolase, a strong inhibitor of DNA methyltransferase and of the methyl donor S-adenosyl-methionine (SAM). HPLC analysis confirmed higher SAM/SAH ratio along with a metabolic reprogramming. Interestingly, an increased LDHA nuclear localization was detected both in mH2A1.1-OE cells and MDS-MSCs, probably depending on MSC inflammatory phenotype. Finally, coculturing healthy mH2A1.1-OE MSCs with CD34+ cells, we found a significant reduction in the number of CD34+ cells, which was reflected in a decreased number of colony forming units (CFU-Cs). These results suggest a key role of mH2A1.1 in driving the crosstalk between epigenetic signaling, inflammation, and cell metabolism networks in MDS-MSCs.


Assuntos
Células-Tronco Mesenquimais , Síndromes Mielodisplásicas , Neoplasias , Humanos , DNA/metabolismo , Epigênese Genética , Histonas/metabolismo , Inflamação/patologia , Células-Tronco Mesenquimais/metabolismo , Síndromes Mielodisplásicas/patologia , Neoplasias/patologia , Proteômica , Receptor 4 Toll-Like/metabolismo , Microambiente Tumoral
2.
Br J Pharmacol ; 172(6): 1557-73, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24641722

RESUMO

Carbon monoxide (CO) is enzymatically generated in mammalian cells alongside the liberation of iron and the production of biliverdin and bilirubin. This occurs during the degradation of haem by haem oxygenase (HO) enzymes, a class of ubiquitous proteins consisting of constitutive and inducible isoforms. The constitutive HO2 is present in the gastrointestinal tract in neurons and interstitial cells of Cajal and CO released from these cells might contribute to intestinal inhibitory neurotransmission and/or to the control of intestinal smooth muscle cell membrane potential. On the other hand, increased expression of the inducible HO1 is now recognized as a beneficial response to oxidative stress and inflammation. Among the products of haem metabolism, CO appears to contribute primarily to the antioxidant and anti-inflammatory effects of the HO1 pathway explaining the studies conducted to exploit CO as a possible therapeutic agent. This article reviews the effects and, as far as known today, the mechanism(s) of action of CO administered either as CO gas or via CO-releasing molecules in acute gastrointestinal inflammation. We provide here a comprehensive overview on the effect of CO in experimental in vivo models of post-operative ileus, intestinal injury during sepsis and necrotizing enterocolitis. In addition, we will analyse the in vitro data obtained so far on the effect of CO on intestinal epithelial cell lines exposed to cytokines, considering the important role of the intestinal mucosa in the pathology of gastrointestinal inflammation.


Assuntos
Monóxido de Carbono/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Inflamação/patologia , Doença Aguda , Animais , Citocinas/metabolismo , Gastroenteropatias/patologia , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/patologia , Humanos , Mucosa Intestinal/patologia , Estresse Oxidativo/fisiologia
3.
Clin Exp Immunol ; 163(3): 368-74, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21235533

RESUMO

We have evaluated the effects of the carbon monoxide-releasing molecule CORM-A1 [Na(2) (BH(3) CO(2) ); ALF421] on the development of relapsing-remitting experimental allergic encephalomyelitis (EAE) in SJL mice, an established model of multiple sclerosis (MS). The data show that the prolonged prophylactic administration of CORM-A1 improves the clinical and histopathological signs of EAE, as shown by a reduced cumulative score, shorter duration and a lower cumulative incidence of the disease as well as milder inflammatory infiltrations of the spinal cords. This study suggests that the use of CORM-A1 might represent a novel therapeutic strategy for the treatment of multiple sclerosis.


Assuntos
Boranos/uso terapêutico , Monóxido de Carbono/uso terapêutico , Carbonatos/uso terapêutico , Encefalomielite Autoimune Experimental/prevenção & controle , Proteína Proteolipídica de Mielina/imunologia , Fragmentos de Peptídeos/imunologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/sangue , Anti-Inflamatórios não Esteroides/farmacologia , Anti-Inflamatórios não Esteroides/uso terapêutico , Peso Corporal/efeitos dos fármacos , Boranos/farmacocinética , Monóxido de Carbono/administração & dosagem , Monóxido de Carbono/sangue , Monóxido de Carbono/farmacologia , Carbonatos/farmacocinética , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Encefalomielite Autoimune Experimental/diagnóstico , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Camundongos , Neutrófilos/patologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/patologia
4.
Cryobiology ; 58(3): 248-55, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19444967

RESUMO

UNLABELLED: There is increasing evidence that carbon monoxide (CO), a signaling molecule generated during the degradation of heme by heme oxygenase-1 (HO-1) in biological systems, has a variety of cytoprotective actions, including anti-hypoxic effects at low temperatures. However, during liver cold preservation, a direct effect needs to be established. Here, we designed a study to analyze the role of CO, delivered via a carbon monoxide-releasing molecule (CO-RM) in the maintenance of liver function, and integrity in rats during cold ischemia/reperfusion (CI/R) injury. We used an isolated normothermic perfused liver system (INPL) following a clinically relevant model of ex vivo 48 h cold ischemia stored in a modified University of Wisconsin (UW) solution, to determine the specific effects of CO in a rat model. CO was generated from 50 microM tricarbonylchloro ruthenium-glycinato (CORM-3), a water-soluble transition metal carbonyl that exerts pharmacological activities via the liberation of controlled amounts of CO in biological systems. The physiological effects of CORM-3 were confirmed by the parallel use of a specific inactive compound (iCORM-3), which does not liberate CO in the cellular environment. CORM-3 addition was found to prevent the injury caused by cold storage by improving significantly the perfusion flow during reperfusion (by almost 90%), and by decreasing the intrahepatic resistance (by 88%) when compared with livers cold preserved in UW alone. Also, CORM-3 supplementation preserved good metabolic capacity as indicated by hepatic oxygen consumption, glycogen content, and release of lactate dehydrogenase. Liver histology was also partially preserved by CORM-3 treatment. CONCLUSIONS: These findings suggest that CO-RM could be utilized as adjuvant therapeutics in UW solutions to limit the injury sustained by donor livers during cold storage prior to transplantation, as has been similarly proposed for the heart, and kidney.


Assuntos
Temperatura Baixa , Fígado , Compostos Organometálicos/farmacologia , Substâncias Protetoras/farmacologia , Preservação de Tecido/métodos , Animais , Monóxido de Carbono/metabolismo , Glicogênio/metabolismo , Lactato Desidrogenases/metabolismo , Fígado/metabolismo , Masculino , Consumo de Oxigênio/fisiologia , Ratos , Ratos Wistar
5.
Gut ; 58(3): 347-56, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19022916

RESUMO

BACKGROUND AND AIMS: Treatment with carbon monoxide (CO) inhalation has been shown to ameliorate postoperative ileus (POI) in rodents and swine. The aim of this study was to investigate whether CO liberated from water-soluble CO-releasing molecules (CO-RMs) can protect against POI in mice and to elucidate the mechanisms involved. METHODS: Ileus was induced by surgical manipulation of the small intestine (IM). Intestinal contractility-transit was evaluated by video-fluorescence imaging. Leucocyte infiltration (myeloperoxidase), inflammatory parameters (ELISA), oxidative stress (lipid peroxidation), and haem oxygenase (HO)/inducible nitric oxide synthase (iNOS) enzyme activity were measured in the intestinal mucosa and muscularis propria. RESULTS: Intestinal contractility and transit were markedly restored when manipulated mice were pre-treated with CO-RMs. Intestinal leucocyte infiltration, expression levels of interleukin 6 (IL6), monocyte chemoattractant protein-1 and intercellular adhesion molecule-1, as well as iNOS activity were reduced by treatment with CORM-3 (a transition metal carbonyl that releases CO very rapidly); whereas expression of IL10/HO-1 was further increased when compared to nontreated manipulated mice. Moreover, treatment with CORM-3 markedly reduced oxidative stress and extracellular signal-related kinase (ERK)1/2 activation in both mucosa (early response) and muscularis (biphasic response). The p38 mitogen-activated protein kinase inhibitor SB203580 abolished CORM-3-mediated HO-1 induction. The HO inhibitor chromium mesoporphyrin only partially reversed the protective effects of CORM-3 on inflammation/oxidative stress in the muscularis, but completely abrogated CORM-3-mediated inhibition of the early "oxidative burst" in the mucosa. CONCLUSIONS: Pre-treatment with CO-RMs markedly reduced IM-induced intestinal muscularis inflammation. These protective effects are, at least in part, mediated through induction of HO-1, in a p38-dependent manner, as well as reduction of ERK1/2 activation. In addition, CORM-induced HO-1 induction reduces the early "oxidative burst" in the mucosa following IM.


Assuntos
Monóxido de Carbono/administração & dosagem , Heme Oxigenase-1/biossíntese , Íleus/prevenção & controle , Intestino Delgado , Estresse Oxidativo/efeitos dos fármacos , Complicações Pós-Operatórias/prevenção & controle , Administração por Inalação , Animais , Carboxihemoglobina/metabolismo , Trânsito Gastrointestinal/fisiologia , Heme Oxigenase-1/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo II/metabolismo , Distribuição Aleatória , Transdução de Sinais
6.
Br J Ophthalmol ; 93(2): 254-7, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18977789

RESUMO

BACKGROUND: Carbon monoxide-releasing molecules (CORMs) are a novel group of substances that are capable of modulating physiological functions via the liberation of CO. AIMS: This study was undertaken to investigate the effects of CORM-3, a water-soluble CO-releasing agent, on two rabbit models of ocular hypertension. METHODS: Ocular hypertension was induced by injecting alpha-chymotrypsin in the rabbit eye. The dose-response effect of CORM-3 on IOP was assessed by topical administration of the drug (0.001, 0.01, 0.1 and 1%). Ocular hypertension was also obtained by weekly subconjunctival injection of betamethasone, and animals were treated topically with CORM-3. A group of animals in both models was treated with the inactive form of the drug (iCORM-3). RESULTS: CORM-3 induced a dose-dependent reduction in IOP in rabbits treated with alpha-chymotrypsin. A similar reduction in IOP was observed in rabbits with betamethasone-induced ocular hypertension treated with the drug. Treatment with the iCORM-3 had no effect on IOP in both models. CONCLUSIONS: Treatment with CORM-3 is associated with a reduction in IOP in two different rabbit models of ocular hypertension. These results support previous findings on the effect of haem oxygenase-derived CO on IOP and suggest a direct involvement of CO system in the regulation of ocular pressure probably through the modulation of aqueous humour dynamics.


Assuntos
Anti-Hipertensivos/uso terapêutico , Hipertensão Ocular/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Animais , Anti-Hipertensivos/administração & dosagem , Quimotripsina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Pressão Intraocular/efeitos dos fármacos , Masculino , Hipertensão Ocular/induzido quimicamente , Hipertensão Ocular/fisiopatologia , Compostos Organometálicos/administração & dosagem , Coelhos
7.
Ann Rheum Dis ; 67(9): 1211-7, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18063671

RESUMO

OBJECTIVE: CO-releasing molecules (CO-RMs) are a novel class of anti-inflammatory agents. We have examined the possible therapeutic effects of CORM-3 in collagen-induced arthritis (CIA). METHODS: Arthritis was induced in DBA-1/J mice by type II collagen. Animals were treated with CORM-3 (5 and 10 mg/kg/day, intraperitoneally) or the inactive compound iCORM-3 (10 mg/kg/day, intraperitoneally) unable to release CO, from days 22 to 31. Production of anti-type II collagen antibodies, cytokines and cartilage olimeric matrix protein (COMP) was evaluated by enzyme-linked immunosorbent assay, and prostaglandin E(2) (PGE(2)) by radioimmunoassay. Localisation of cyclooxygenase-2 (COX-2), haem oxygenase-1 (HO-1), intercellular adhesion molecule-1 (ICAM-1) and receptor activator of nuclear factor kappaB ligand (RANKL) was examined by immunohistochemistry. RESULTS: Therapeutic administration of CORM-3 suppressed clinical and histopathological manifestations of disease. The levels of PGE(2), interleukin (IL)1beta, IL2, IL6, IL10 and tumour necrosis factor (TNF)alpha in joint tissues were inhibited by CORM-3. By contrast, CORM-3 augmented IL4. Anti-type II collagen antibodies and COMP levels in serum were reduced by CORM-3. Treatment with CORM-3 decreased cellular infiltration, joint inflammation and destruction, as well as the expression of COX-2, ICAM-1 and RANKL, whereas HO-1 increased. These beneficial effects were due to CO release, as iCORM-3 was ineffective. CONCLUSION: This study reveals the antiarthritic properties of CORM-3 in the CIA model and supports the notion that CO-RMs could be developed as a novel strategy for the treatment of inflammatory and arthritic conditions.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Compostos Organometálicos/uso terapêutico , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Ciclo-Oxigenase 2/metabolismo , Citocinas/biossíntese , Dinoprostona/biossíntese , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Heme Oxigenase-1/metabolismo , Mediadores da Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Ligante RANK/metabolismo
8.
Biochem Soc Trans ; 35(Pt 5): 1142-6, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17956297

RESUMO

The well-known adverse effects of CO (carbon monoxide) intoxication are counterbalanced by its positive actions when small amounts are produced intracellularly by the cytoprotective enzyme HO-1 (haem oxygenase-1). As compelling scientific evidence accumulated to sustain that HO-1 plays a fundamental role in counteracting vascular and inflammatory disorders, we began to appreciate that a controlled delivery of CO to mammals may provide therapeutic benefits in a number of pathological states. This is the rationale for the recent development of CO-RMs (CO-releasing molecules), a group of compounds capable of carrying and liberating controlled quantities of CO in cellular systems, which offer a plausible tool for studying the pharmacological effects of this gas and identifying its mechanism(s) of action. The present review will highlight the encouraging results obtained so far on the vasodilatory, anti-ischaemic and anti-inflammatory effects elicited by CO-RMs in in vitro and in vivo models with an emphasis on the prospect of converting chemical CO carriers into CO-based pharmaceuticals.


Assuntos
Monóxido de Carbono/metabolismo , Inflamação/prevenção & controle , Isquemia/prevenção & controle , Vasodilatação/efeitos dos fármacos , Humanos , Lipídeos/química , Solubilidade
10.
Br J Pharmacol ; 149(8): 1104-12, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17057755

RESUMO

BACKGROUND AND PURPOSE: Carbon monoxide (CO) generated by the enzyme haeme oxygenase-1 (HO-1) during the breakdown of haeme is known to mediate a number of biological effects. Here, we investigated whether CO liberated from two water soluble carbon monoxide-releasing molecules (CO-RMs) exerts inotropic effects on the myocardium. EXPERIMENTAL APPROACH: Rat isolated hearts perfused either at constant flow or constant pressure were used to test the effect of CO-RMs. KEY RESULTS: CORM-3, a fast CO releaser, produced a direct positive inotropic effect when cumulative doses (3, 10 and 30 microg min(-1)) or a single dose (5 microM) were infused at either constant coronary pressure (CCP) or constant coronary flow (CCF). The inotropic effect mediated by CORM-3 was abolished by blockade of soluble guanylate cyclase or Na(+)/H(+) exchanger, but not by inhibitors of L-type Ca(2+) channels and protein kinase C. CORM-3 also caused a slight reduction in heart rate but did not alter coronary flow. In contrast, the slow CO releaser CORM-A1 produced significant coronary vasodilatation when given at the highest concentration (30 mug min(-1)) but exerted no effect on myocardial contractility or heart rate. CONCLUSION AND IMPLICATIONS: A rapid CO release from CORM-3 exerts a direct positive inotropic effect on rat isolated perfused hearts, whereas CO slowly released by CORM-A1 had no effect on myocardial contractility but caused significant coronary vasodilatation. Both cGMP and Na(+)/H(+) exchange appear to be involved in this effect but further work is needed to determine the relative contribution of each pathway in CO-mediated inotropic effect.


Assuntos
Boranos/farmacologia , Monóxido de Carbono/metabolismo , Carbonatos/farmacologia , Cardiotônicos/farmacologia , Miocárdio/metabolismo , Compostos Organometálicos/farmacologia , Alcaloides/farmacologia , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Benzofenantridinas/farmacologia , Boranos/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Carbonatos/metabolismo , Inibidores Enzimáticos/farmacologia , Coração/efeitos dos fármacos , Cinética , Masculino , Nifedipino/farmacologia , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais/efeitos dos fármacos
11.
Nephron Exp Nephrol ; 104(4): e135-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16902317

RESUMO

Heme oxygenase (HO) isoforms catalyze the conversion of heme to carbon monoxide (CO) and biliverdin/bilirubin with a concurrent release of iron. There is strong evidence that HO activity and products play a major role in renoprotection, however the exact molecular mechanisms underlying the beneficial effects exerted by this pathway are not fully understood. This review is aimed at illustrating the possible mechanism/s by which HO is renoprotective in the context of ischemia/reperfusion. We will first analyze the effects of exogenous administration of bilirubin/biliverdin and CO and then describe their biological activities once generated endogenously following stimulation of the HO pathway by either pharmacological means or gene targeting-mediated approaches.


Assuntos
Biliverdina/fisiologia , Monóxido de Carbono/fisiologia , Heme Oxigenase (Desciclizante)/metabolismo , Rim/irrigação sanguínea , Traumatismo por Reperfusão/fisiopatologia , Animais , Bilirrubina/fisiologia , Humanos
13.
Kidney Int ; 69(2): 239-47, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16408112

RESUMO

Low concentrations of carbon monoxide (CO) can protect tissues against ischemia-reperfusion (I-R) injury. We have recently identified a novel class of compounds, CO-releasing molecules (CO-RMs), which exert important pharmacological activities by carrying and delivering CO to biological systems. Here, we examined the possible beneficial effects of CO liberated from CO-RMs on the damage inflicted by cold storage and I-R in isolated perfused kidneys. Hemodynamic and biochemical parameters as well as mitochondrial respiration were measured in isolated perfused rabbit kidneys that were previously flushed with CO-RMs and stored at 4 degrees C for 24 h. Two water-soluble CO-RMs were tested: (1) sodium boranocarbonate (CORM-A1), a boron-containing carbonate that releases CO at a slow rate, and (2) tricarbonylchloro(glycinato)ruthenium(II) (CORM-3), a transition metal carbonyl that liberates CO very rapidly in solution. Kidneys flushed with Celsior solution supplemented with CO-RMs (50 microM) and stored at 4 degrees C for 24 h displayed at reperfusion a significantly higher perfusion flow rate (PFR), glomerular filtration rate, and sodium and glucose reabsorption rates compared to control kidneys flushed with Celsior solution alone. Addition of 1H-[1,2,4]oxadiazolo[4,3-alpha]quinoxalin-1-one (ODQ), a guanylate cyclase inhibitor, prevented the increase in PFR mediated by CO-RMs. The respiratory control index from kidney mitochondria treated with CO-RMs was also markedly increased. Notably, renal protection was lost when kidneys were flushed with Celsior containing an inactive compound (iCO-RM), which had been deliberately depleted of CO. CO-RMs are effective therapeutic agents that deliver CO during kidney cold preservation and can be used to ameliorate vascular activity, energy metabolism and renal function at reperfusion.


Assuntos
Monóxido de Carbono/farmacologia , Rim/efeitos dos fármacos , Preservação de Órgãos , Traumatismo por Reperfusão/prevenção & controle , Animais , Monóxido de Carbono/metabolismo , Temperatura Baixa , Rim/irrigação sanguínea , Rim/fisiologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxidiazóis/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Quinoxalinas/farmacologia , Coelhos
15.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 403-8, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16309591

RESUMO

Matrix metalloproteinases (MMPs), particularly MMP-1 and MMP-2, are involved in the pathophysiology of emphysema. MMPs contain zinc in the catalytic site and its expression is regulated transcriptionally via mitogen activated protein kinases (MAPKs). Carbon monoxide (CO), one of the end products of heme oxygenase activity, has anti-inflammatory properties, which are mediated, at least in part, by activation of p38 MAPK. Furthermore, CO has the unique ability to bind to metal centers in proteins and can affect their specific activity. Therefore, we hypothesized that CO could inhibit MMPs expression and/or activity. Here we show that a recently identified carbon monoxide-releasing molecule, [Ru(CO)3Cl2]2 (or CORM-2) inhibits MMP-1 and MMP-2 mRNA expression in the human lung epithelial cell line A549. The MMPs mRNA expression was unaffected by the p38 MAPK inhibitor SB203580, but in the case of MMP-1 was reversed by the antioxidant N-acetylcysteine. In addition, CORM-2 inhibited both MMP-1 and MMP-2 activities. Interestingly, no effect was observed with (Ru(DMSO)4Cl2), a negative control that does not contain CO groups. To the best of our knowledge this is the first evidence on the effect of CO on MMPs expression and activity. This effect could have important implications in the pathophysiology of emphysema and other diseases involving proteases/antiproteases imbalance.


Assuntos
Monóxido de Carbono/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Alvéolos Pulmonares/citologia , Linhagem Celular Tumoral , Humanos , Interleucina-1beta/farmacologia , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Compostos Organometálicos/farmacologia , RNA Mensageiro/genética
16.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 409-23, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16309592

RESUMO

The inducible stress protein heme oxygenase-1 (HO-1) has been linked to tissue and organ protection against the deleterious actions of many pathological conditions, including endotoxin challenge. Similar protection can be achieved by the main products of heme oxygenase activity, namely bilirubin and carbon monoxide (CO). Since the identification of novel chemical compounds that liberate CO in biological systems (CO-releasing molecules or CO-RMs), our group and others have had access to a convenient and simple pharmacological tool that enables to study the role of CO in physiological functions. This article will review the scientific literature published to date on CO-RMs, with emphasis on the in vitro, ex vivo and in vivo experimental models employed to determine the contribution of CO to cellular mechanisms. In addition, we will report on the effect of heme oxygenase-related substances, such as bilirubin, CORM-3 and hemin, in a model of endotoxin-induced hypotension. Among the three different approaches examined, CORM-3 proved the most effective agent in reducing the fall in blood pressure caused by endotoxin. Furthermore, heme oxygenase-related substances affected the endotoxin-stimulated induction and distribution of hepatic HO-1 and inducible nitric oxide synthase (iNOS). Thus, it emerges that CO-RMs could exert important biological actions in the context of endotoxic-mediated dysfunction.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Monóxido de Carbono/metabolismo , Endotoxinas/farmacologia , Hipotensão/induzido quimicamente , Hipotensão/fisiopatologia , Animais , Regulação Enzimológica da Expressão Gênica/genética , Coração/efeitos dos fármacos , Heme Oxigenase-1/genética , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Miocárdio/enzimologia , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
17.
Cell Mol Biol (Noisy-le-grand) ; 51(4): 425-32, 2005 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-16309593

RESUMO

Emerging evidence reveals that heme oxygenase-1 (HO-1) and its product carbon monoxide (CO) can exert diverse biological and cytoprotective effects. Our group has recently identified a new class of compounds (CO-releasing molecules or CO-RMs) that can carry and deliver CO to biological systems and can be used to examine the physiological properties of CO. Here, we evaluated the influence of endogenously-generated CO (via HO-1 induction by hemin) and CO liberated from exogenously supplied CO-RMs on mitochondrial function. Renal mitochondria were isolated either from rats with increased HO-1 or from untreated animals, the latter being exposed to different concentrations of CO-RMs (10-100 microM). We found that mitochondrial oxygen uptake was significantly reduced in kidneys after HO-1 induction and, in a similar fashion, CO-RMs inhibited mitochondrial function in a concentration-dependent manner. Specifically, a marked depression of state 3 was observed resulting in a significant decrease in respiratory control index (RCI) values. When mitochondria were incubated with the inactive forms of CO-RMs, which are devoid of CO, the respiratory parameters remained unchanged. In summary, the results indicate that HO-1 induction and enhanced CO decrease renal oxygen consumption and alter mitochondrial function suggesting that CO could be a physiological regulator of mitochondrial oxidative phosphorylation.


Assuntos
Monóxido de Carbono/metabolismo , Respiração Celular/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Animais , Heme Oxigenase (Desciclizante)/metabolismo , Hemina/farmacologia , Masculino , Estrutura Molecular , Ratos , Ratos Sprague-Dawley
18.
Mol Pharmacol ; 61(3): 554-61, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11854435

RESUMO

Heme oxygenase-1 (HO-1) is a redox-sensitive inducible protein that provides efficient cytoprotection against oxidative stress. Curcumin, a polyphenolic natural compound that possesses anti-tumor and anti-inflammatory properties, has been reported recently to induce potently HO-1 expression in vascular endothelial cells (Free Rad Biol Med 28:1303-1312, 2000). Here, we extend our previous findings by showing that caffeic acid phenethyl ester (CAPE), another plant-derived phenolic agent, markedly increases heme oxygenase activity and HO-1 protein in astrocytes. The effect seems to be related to the peculiar chemical structures of curcumin and CAPE, because analogous antioxidants containing only portions of these two molecules were totally ineffective. At a final concentration of 30 microM, both curcumin and CAPE maximally up-regulated heme oxygenase activity while promoting marked cytotoxicity at higher concentrations (50-100 microM). Similar results were obtained with Curcumin-95, a mixture of curcuminoids commonly used as a dietary supplement. Incubation of astrocytes with curcumin or CAPE at concentrations that promoted maximal heme oxygenase activity resulted in an early increase in reduced glutathione followed by a significant elevation in oxidized glutathione contents. A curcumin-mediated increase in heme oxygenase activity was not affected by the glutathione precursor and thiol donor N-acetyl-L-cysteine. These data suggest that regulation of HO-1 expression by polyphenolic compounds is evoked by a distinctive mechanism which is not necessarily linked to changes in glutathione but might depend on redox signals sustained by specific and targeted sulfydryl groups. This study identifies a novel class of natural substances that could be used for therapeutic purposes as potent inducers of HO-1 in the protection of tissues against inflammatory and neurodegenerative conditions.


Assuntos
Astrócitos/efeitos dos fármacos , Ácidos Cafeicos/farmacologia , Curcumina/farmacologia , Heme Oxigenase (Desciclizante)/biossíntese , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/farmacologia , Acetilcisteína/farmacologia , Animais , Astrócitos/enzimologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Glutationa/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Heme Oxigenase-1 , Ratos , Regulação para Cima/efeitos dos fármacos
19.
Cell Mol Biol (Noisy-le-grand) ; 48(8): 885-94, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12699247

RESUMO

Heme oxygenase-1 (HO-1) is a cytoprotective enzyme, the expression of which is highly sensitive to induction by pro-oxidant stimuli including the substrate heme and reactive oxygen species. Conceptually, the perception that HO-1 plays a key role in response to oxidative damage is paralleled by evidence showing high expression of HO-1 in a variety of cell systems challenged with nitric oxide (NO) or NO-derivatives, thus revealing a potential biological function for HO-1 against nitrosative stress. In this study, we report that exposure of cardiac cells to hemin (5-20 microM) in combination with compounds that liberate nitroxyl (HNO/NO-) or release NO significantly potentiates HO-1 mRNA and protein expression leading to a remarkable increase in heme oxygenase activity under both normoxic and hypoxic conditions. The amplification of the heme oxygenase pathway appears to involve a direct interaction between heme and the NO groups, as the ability of both NO(-)- and NO-releasing agents to induce HO-1 is totally lost by their pre-incubation for 1 hr in complete medium prior to cell treatment but is highly preserved by addition of hemin during the preincubation step. In addition, we show that the redox-sensitive transcription factor Nrf2 is highly expressed in the nuclear fraction of cells exposed to the NO- generator and that this effect is totally abolished by the presence of N-acetyl-L-cysteine. Interestingly, the expression of Nrf2 is gradually intensified by treating cells with a combination of the NO- releaser and increasing concentrations of hemin. Thus, a strict parallelism exists between the extent of HO-1 induction and expression of Nrf2 elicited by the heme-NO interaction. We propose that modification of the iron protoporphyrin centers by NO groups to modulate HO-1 expression might be regarded as a molecular switch to maximize heme oxygenase enzymatic activity and consequently mitigate the redox imbalance imposed by oxidative and nitrosative stress.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Heme/metabolismo , Óxido Nítrico/metabolismo , Transativadores/metabolismo , Acetilcisteína/farmacologia , Animais , Ânions , Western Blotting , Linhagem Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Heme/química , Heme Oxigenase-1 , Hemina/química , Miocárdio/citologia , Fator 2 Relacionado a NF-E2 , Óxido Nítrico/química , Nitrogênio/metabolismo , Oxidantes/farmacologia , Oxirredução , Estresse Oxidativo , Oxigênio/metabolismo , Ligação Proteica , RNA/metabolismo , RNA Mensageiro/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Compostos de Sulfidrila/metabolismo , Temperatura , Fatores de Tempo
20.
FEBS Lett ; 508(3): 403-6, 2001 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-11728461

RESUMO

The disrupted metabolism of homocysteine (Hcy) causes hyperhomocysteinemia, a condition associated with the impairment of nitric oxide (NO) bio-availability, tissue hypoxia and increased risk of vascular disease. Here, we examined how Hcy modulates the induction of the stress protein haem oxygenase-1 (HO-1) evoked by NO releasing agents and hypoxia in vascular endothelial cells. We found that Hcy (0.5 mM) markedly reduced the increase in haem oxygenase activity and HO-1 protein expression induced by sodium nitroprusside (SNP, 0.5 mM) but did not affect HO-1 activation mediated by S-nitroso-N-acetyl-penicillamine. Cells pre-treated with Hcy followed by addition of fresh medium containing SNP still exhibited an augmented haem oxygenase activity. Interestingly, high levels of Hcy were also able to abolish hypoxia-mediated HO-1 expression in a concentration-dependent manner. These novel findings indicate that hyperhomocysteinemia interferes with crucial signaling pathways required by cells to respond and adapt to stressful conditions.


Assuntos
Hipóxia Celular , Endotélio Vascular/enzimologia , Heme Oxigenase (Desciclizante)/biossíntese , Homocisteína/farmacologia , Óxido Nítrico/metabolismo , Animais , Bovinos , Células Cultivadas , Meios de Cultura , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Indução Enzimática/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Nitroprussiato/farmacologia , S-Nitroso-N-Acetilpenicilamina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...