Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Tissue Eng Regen Med ; 14(6): 807-818, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32330363

RESUMO

Mounting evidence showing that local nitric oxide (NO) delivery may significantly improve the wound healing process has stimulated the development of wound dressings capable of releasing NO topically. Herein, we describe the preparation of a self-expandable NO-releasing hydrolyzed collagen sponge (CS), charged with the endogenously found NO donor, S-nitrosoglutathione (GSNO). We show that cold pressed and GSNO-charged CS (CS/GSNO) undergo self-expansion to its original 3D shape upon water absorption to a swelling degree of 2,300 wt%, triggering the release of free NO. Topical application of compressed CS/GSNO on wounds in an animal model showed that exudate absorption by CS/GSNO leads to the release of higher NO doses during the inflammatory phase and progressively lower NO doses at later stages of the healing process. Moreover, treated animals showed significant increase in the mRNA expression levels of monocyte chemoattractant protein-1 (MCP-1), murine macrophage marker (F4/80), transforming growth factor beta (TGF-ß), stromal cell-derived factor 1 (SDF-1), insulin-like growth factor-1 (IGF-1), nitric oxide synthase(iNOS), and matrix metalloproteinase(MMP-9). Cluster differentiation 31 (CD31), vascular endothelial growth factor (VEGF), and F4/80 were measured on Days 7 and 12 by immunohistochemistry in the cicatricial tissue. These results indicate that the topical delivery of NO enhances the migration and infiltration of leucocytes, macrophages, and keratinocytes to the wounded tissue, as well as the neovascularization and collagen deposition, which are correlated with an accelerated wound closure. Thus, self-expandable CS/GSNO may represent a novel biocompatible and active wound dress for the topical delivery of NO on wounds.


Assuntos
Colágeno , Óxido Nítrico , S-Nitrosoglutationa , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões , Animais , Colágeno/química , Colágeno/farmacologia , Modelos Animais de Doenças , Implantes de Medicamento/química , Implantes de Medicamento/farmacocinética , Implantes de Medicamento/farmacologia , Masculino , Camundongos , Óxido Nítrico/química , Óxido Nítrico/farmacocinética , Óxido Nítrico/farmacologia , S-Nitrosoglutationa/química , S-Nitrosoglutationa/farmacocinética , S-Nitrosoglutationa/farmacologia , Ferimentos e Lesões/tratamento farmacológico , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
2.
Drug Dev Ind Pharm ; 45(3): 423-429, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30449192

RESUMO

OBJECTIVE: S-nitrosogluthatione (GSNO), a S-nitrosothiol, is a commonly used as nitric oxide (NO•) donor. However, its half-life is too short for a direct therapeutic use. To protect and ensure a sustained release of NO•, the encapsulation of GSNO into nanoparticles may be an interesting option. METHODS: In this work, we have investigated the early (4 h) and late (24 h) transcriptomic response of THP-1 human monocytes cells to two doses (1.4 and 6 µM) of either free or Eudragit® nano-encapsulated GSNO using RNA microarray. RESULTS: After exposure to free GSNO, genes mainly involved in apoptosis, cell differentiation, immune response and metabolic processes were differentially expressed. Although, cells exposed to free or encapsulated GSNO behave differently, activation of genes involved in blood coagulation, immune response and cell cycle was observed in both conditions. CONCLUSIONS: These results suggest that the encapsulation of low doses of GSNO into Eudragit® nanoparticles leads to a progressive release of GSNO making this compound a possible oral therapy for several biomedical applications like inflammatory bowel diseases.


Assuntos
S-Nitrosoglutationa/farmacocinética , Transcriptoma/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Meia-Vida , Humanos , Imunidade/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Nanopartículas/metabolismo , Óxido Nítrico/metabolismo , Ácidos Polimetacrílicos/química , Células THP-1
3.
Rapid Commun Mass Spectrom ; 33(1): 1-11, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30248720

RESUMO

RATIONALE: The potency of S-nitrosoglutathione (GSNO) as a nitric oxide (NO) donor to treat cardiovascular diseases (CVDs) has been highlighted in numerous studies. In order to study its bioavailability after oral administration, which represents the most convenient route for the chronic treatment of CVDs, it is essential to develop an analytical method permitting (i) the simultaneous measurement of GSNO metabolites, i.e. nitrite, S-nitrosothiols (RSNOs) and nitrate and (ii) to distinguish them from other sources (endogenous synthesis and diet). METHODS: Exogenous GSNO was labeled with 15 N, and the GS15 NO metabolites after conversion into the nitrite ion were derivatized with 2,3-diaminonaphthalene. The resulting 2,3-naphthotriazole was quantified by liquid chromatography/tandem ion trap mass spectrometry (LC/ITMS/MS) in multiple reaction monitoring mode after Higher-energy Collision-induced Dissociation (HCD). Finally, the validated method was applied to an in vitro model of the intestinal barrier (monolayer of Caco-2 cells) to study GS15 NO intestinal permeability. RESULTS: A LC/ITMS/MS method based on an original transition (m/z 171 to 156) for sodium 15 N-nitrite, GS15 NO and sodium 15 N-nitrate measurements was validated, with recoveries of 100.8 ± 3.8, 98.0 ± 2.7 and 104.1 ± 3.3%, respectively. Intra- and inter-day variabilities were below 13.4 and 12.6%, and the limit of quantification reached 5 nM (signal over blank = 4). The permeability of labeled GS15 NO (10-100 µM) was evaluated by calculating its apparent permeability coefficient (Papp ). CONCLUSIONS: A quantitative LC/ITMS/MS method using HCD was developed for the first time to selectively monitor GS15 NO metabolites. The assay allowed evaluation of GS15 NO intestinal permeability and situated this drug candidate within the middle permeability class according to FDA guidelines. In addition, the present method has opened the perspective of a more fundamental work aiming at studying the fragmentation mechanism leading to the ion at m/z 156 in HCD tandem mass spectrometry in the presence of acetonitrile.


Assuntos
Cromatografia Líquida/métodos , Óxido Nítrico/análise , Óxido Nítrico/metabolismo , S-Nitrosoglutationa/farmacocinética , Espectrometria de Massas em Tandem/métodos , 2-Naftilamina/análogos & derivados , 2-Naftilamina/química , Células CACO-2 , Humanos , Absorção Intestinal/efeitos dos fármacos , Limite de Detecção , Nitritos/química , Reprodutibilidade dos Testes , S-Nitrosoglutationa/metabolismo , Espectrometria de Massas em Tandem/instrumentação
4.
Acta Biomater ; 74: 312-325, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29777958

RESUMO

Topical nitric oxide (NO) delivery has been shown to accelerate wound healing. However, delivering NO to wounds at appropriate rates and doses requires new biomaterial-based strategies. Here, we describe the development of supramolecular interpolymer complex hydrogels comprising PEO-PPO-PEO (F127) micelles embedded in a poly(acrylic acid) (PAA) matrix, with S-nitrosoglutathione (GSNO) molecules dissolved in the hydrophilic domain. We show that PAA:F127/GSNO hydrogels start releasing NO upon hydration at rates controlled by their rates of water absorption. SAXS measurements indicate that the supramolecular structure of the hydrogels retains long-range order domains of F127 micelles. The PAA/F1227 hydrogels displayed dense morphologies and reduced rates of hydration. The NO release rates remain constant over the first 200 min, are directly correlated with the hydration rates of the PAA:F127/GSNO hydrogels, and can be modulated in the range of 40 nmol/g h to 1.5 µmol/g h by changing the PAA:F127 mass ratio. Long-term NO-release profiles over 5 days are governed by the first-order exponential decay of GSNO, with half-lives in the range of 0.5-3.4 days. A preliminary in vivo study on full-thickness excisional wounds in mice showed that topical NO release from the PAA:F127/GSNO hydrogels is triggered by exudate absorption and leads to increased angiogenesis and collagen fiber organization, as well as TGF-ß, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue. In summary, these results suggest that hydration-controlled NO release from topical PAA:F127/GSNO hydrogels is a potential strategy for enhancing wound healing. STATEMENT OF SIGNIFICANCE: The topical delivery of nitric oxide (NO) to wounds may provide significant beneficial results and represent a promising strategy to treat chronic wounds. However, wound dressings capable of releasing NO after application and allowing the modulation of NO release rates, demand new platforms. Here, we describe a novel strategy to overcome these challenges, based on the use of supramolecular poly(acrylic acid) (PAA):F127 hydrogels charged with the NO donor S-nitrosoglutathione (GSNO) from whereby the NO release can be triggered by exudate absorption and delivered to the wound at rates controlled by the PAA:F127 mass ratio. Preliminary in vivo results offer a proof of concept for this strategy by demonstrating increased angiogenesis; collagen fibers organization; and TGF-ß, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue after topical treatment with a PAA:F127/GSNO hydrogel.


Assuntos
Resinas Acrílicas , Hidrogéis , Óxido Nítrico , Polietilenos , Polipropilenos , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões , Resinas Acrílicas/farmacocinética , Resinas Acrílicas/farmacologia , Animais , Citocinas/biossíntese , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Preparações de Ação Retardada/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hidrogéis/química , Hidrogéis/farmacocinética , Hidrogéis/farmacologia , Camundongos , Micelas , Óxido Nítrico/química , Óxido Nítrico/farmacocinética , Óxido Nítrico/farmacologia , Polietilenos/química , Polietilenos/farmacocinética , Polietilenos/farmacologia , Polipropilenos/química , Polipropilenos/farmacocinética , Polipropilenos/farmacologia , S-Nitrosoglutationa/química , S-Nitrosoglutationa/farmacocinética , S-Nitrosoglutationa/farmacologia , Ferimentos e Lesões/tratamento farmacológico , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
5.
Nitric Oxide ; 59: 1-9, 2016 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-27350118

RESUMO

S-nitrosophytochelatins (SNOPCs) are novel analogues of S-nitrosoglutathione (GSNO) with the advantage of carrying varying ratios of S-nitrosothiol (SNO) moieties per molecule. Our aim was to investigate the in vivo pharmacological potency and biodistribution of these new GSNO analogues after intravenous (i.v.) and intranasal (i.n.) administration in mice. SNOPCs with either two or six SNO groups and GSNO were synthesized and characterized for purity. Compounds were administered i.v. or i.n. at 1 µmol NO/kg body weight to CD-1 mice. Blood pressure was measured and biodistribution studies of total nitrate and nitrite species (NOx) and phytochelatins were performed after i.v. administration. At equivalent doses of NO, it was observed that SNOPC-6 generated a rapid and significantly greater reduction in blood pressure (∼60% reduction compared to saline) whereas GSNO and SNOPC-2 only achieved a 30-35% decrease. The reduction in blood pressure was transient and recovered to baseline levels within ∼2 min for all compounds. NOx species were transiently elevated (over 5 min) in the plasma, lung, heart and liver. Interestingly, a size-dependent phytochelatin accumulation was observed in several tissues including the heart, lungs, kidney, brain and liver. Biodistribution profiles of NOx were also obtained after i.n. administration, showing significant lung retention of NOx over 15 min with minor systemic increases observed from 5 to 15 min. In summary, this study has revealed interesting in vivo pharmacological properties of SNOPCs, with regard to their dramatic hypotensive effects and differing biodistribution patterns following two different routes of administration.


Assuntos
Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/farmacologia , Fitoquelatinas/administração & dosagem , Fitoquelatinas/farmacologia , S-Nitrosotióis/administração & dosagem , S-Nitrosotióis/farmacologia , Administração Intranasal , Administração Intravenosa , Animais , Anti-Hipertensivos/análise , Anti-Hipertensivos/farmacocinética , Pressão Arterial/efeitos dos fármacos , Masculino , Camundongos , Nitratos/análise , Nitritos/análise , Fitoquelatinas/farmacocinética , S-Nitrosoglutationa/farmacocinética , S-Nitrosotióis/análise , S-Nitrosotióis/farmacocinética , Umbeliferonas/análise
6.
Nanomedicine ; 12(7): 1795-1803, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27184095

RESUMO

Alginate/chitosan nanocomposite particles (GSNO-acNCPs), i.e. S-nitrosoglutathione (GSNO) loaded polymeric nanoparticles incorporated into an alginate and chitosan matrix, were developed to increase the effective GSNO loading capacity, a nitric oxide (NO) donor, and to sustain its release from the intestine following oral administration. Compared with free GSNO and GSNO loaded nanoparticles, GSNO-acNCPs promoted 2.7-fold GSNO permeation through a model of intestinal barrier (Caco-2 cells). After oral administration to Wistar rats, GSNO-acNCPs promoted NO storage into the aorta during at least 17h, as highlighted by (i) a long-lasting hyporeactivity to phenylephrine (decrease in maximum vasoconstrictive effect of aortic rings) and (ii) N-acetylcysteine (a thiol which can displace NO from tissues)-induced vasodilation of aorxxtic rings preconstricted with phenylephrine. In conclusion, GSNO-acNCPs enhance GSNO intestinal absorption and promote the formation of releasable NO stores into the rat aorta. GSNO-acNCPs are promising carriers for chronic oral application devoted to the treatment of cardiovascular diseases.


Assuntos
Nanocompostos , Óxido Nítrico/metabolismo , Polímeros , S-Nitrosoglutationa/farmacocinética , Animais , Aorta , Células CACO-2 , Humanos , Absorção Intestinal , Ratos , Ratos Wistar
7.
J Biomed Mater Res A ; 103(10): 3303-11, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25809572

RESUMO

Fabrication of scaffolds loaded with nitric oxide (NO) donors (S-nitrosoglutathione, GSNO, and isosorbide mononitrate, ISMN) with suitable cell compatibility and optimized properties for tissue-engineering applications is reported using "in situ" technique. Based on FDA-approved polymer, solvent and dosage forms, this gentle process allowed the incorporation of the GSNO labile drug into scaffolds made of either poly(lactide-co-glycolide) (PLGA) or PLGA/poly(ɛ-caprolactone) (PCL) blend. During scaffolds manufacturing process including washing cycles, NO donors were leached from scaffolds. However, GSNO and ISMN concentrations in the last washing medium (10(-7) M and 10(-4) M, respectively) were in the range of cell suitability for tissue engineering. Further morphological analyses indicated that smoother surfaces with fewer but bigger pores (compatible with cell penetration and ingrowth) were obtained with PLGA in comparison with PLGA/PCL scaffolds. Among all tested matrices, only unloaded PLGA and GSNO-loaded PLGA/PCL exhibited intermediate cell anchorage, with mitochondrial activity close to the control and an increase in protein content, a prognostic for scaffold cell colonization, defining them as promising candidates. Deeper analyses of these two scaffolds looking at intracellular redox balance through reactive oxygen species production, glutathione, S-nitrosothiols, and nitrite ions content exhibited GSNO-loaded PLGA/PCL as the best of all tested 3D scaffolds for tissue engineering.


Assuntos
Implantes de Medicamento , Miócitos de Músculo Liso/metabolismo , Óxido Nítrico , S-Nitrosoglutationa , Alicerces Teciduais/química , Animais , Implantes de Medicamento/química , Implantes de Medicamento/farmacocinética , Implantes de Medicamento/farmacologia , Ácido Láctico/química , Ácido Láctico/farmacocinética , Ácido Láctico/farmacologia , Miócitos de Músculo Liso/citologia , Óxido Nítrico/química , Óxido Nítrico/farmacocinética , Óxido Nítrico/farmacologia , Poliésteres/química , Poliésteres/farmacocinética , Poliésteres/farmacologia , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacocinética , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Ratos , S-Nitrosoglutationa/química , S-Nitrosoglutationa/farmacocinética , S-Nitrosoglutationa/farmacologia
8.
Colloids Surf B Biointerfaces ; 108: 178-84, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23545088

RESUMO

Clinical complications of implantable polypropylene (PP) meshes used to repair urinary incontinence and vaginal prolapse may be associated with their low surface energy and consequent poor tissue integration. For improving tissue integration, we impregnated monofilament PP meshes with physically crosslinked poly(vinyl alcohol) (PVA), resulting in PVA deposits tightly attached inside the knot spaces of the PP knit. While preserving the mesh porosity, the PVA deposits acted as an array of hydrophilic regions leading to a great increase in the overall mesh wettability, reflected by a contact angle decrease from 111 to ca. 66°. The PVA deposits were also used as reservoirs for the local release of S-nitrosoglutathione (GSNO), a nitric oxide (NO) donor. Plain and impregnated PP meshes (1.0cm×1.0cm) were implanted in the subcutaneous tissue of 21 adult female Wistar rats. Histological analysis of the abdominal wall 21 days after the surgeries revealed lower edema and greater angiogenesis while a marked decrement of NOx concentration in the tissue surrounding the impregnated meshes was observed after 2 days. These results indicate that PVA and PVA/GSNO impregnation might be a new strategy for decreasing the frequency of mesh extrusion after PP mesh implants.


Assuntos
Parede Abdominal/irrigação sanguínea , Materiais Biocompatíveis/química , Doadores de Óxido Nítrico/química , Polipropilenos/química , Álcool de Polivinil/química , S-Nitrosoglutationa/química , Parede Abdominal/cirurgia , Animais , Materiais Biocompatíveis/farmacologia , Feminino , Microscopia Eletrônica de Varredura , Neovascularização Fisiológica , Óxido Nítrico/metabolismo , Álcool de Polivinil/farmacocinética , Ratos , Ratos Wistar , S-Nitrosoglutationa/farmacocinética , Telas Cirúrgicas , Molhabilidade
9.
Kardiologiia ; 51(11): 28-37, 2011.
Artigo em Russo | MEDLINE | ID: mdl-22117768

RESUMO

On the basis of earlier executed studies of hypotensive effect of dinitrosyl iron complexes (DNIC) with glutathione, the drug has been created in industrial conditions named oxacom. Preliminary pharmacological studies of oxacom have not revealed negative qualities. The drug has been now tested in 14 healthy men in whom at single intravenous introduction it caused typical response - a decrease of diastolic as well as systolic arterial pressure on 24-27 mmHg through 3-4 min with subsequent very slow restoration in 8-10 hours. The heart rate after initial rise was quickly normalized. Echocardiography revealed unaltered cardiac output in spite of reduced cardiac filling by 28%. The multilateral analysis of clinical and biochemical data has revealed an absence of essential alterations which could lead to pathological consequences. The drug is recommended for carrying out of the second phase of clinical trial. The comparative study of the efficiency of hypotensive action of oxacom, S-nitrosoglutathione (GS-NO) and sodium nitrite (NO2) in rats has shown that the duration of effect was the greatest at oxacom action.


Assuntos
Pressão Sanguínea/efeitos dos fármacos , Glutationa , Hipertensão/tratamento farmacológico , Ferro , Óxidos de Nitrogênio , S-Nitrosoglutationa/farmacocinética , Nitrito de Sódio/farmacocinética , Adulto , Animais , Disponibilidade Biológica , Avaliação Pré-Clínica de Medicamentos/métodos , Monitoramento de Medicamentos/métodos , Glutationa/administração & dosagem , Glutationa/efeitos adversos , Glutationa/farmacocinética , Glutationa/farmacologia , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Hipotensão/induzido quimicamente , Infusões Intravenosas , Ferro/administração & dosagem , Ferro/efeitos adversos , Ferro/farmacocinética , Ferro/farmacologia , Masculino , Óxido Nítrico/metabolismo , Óxidos de Nitrogênio/administração & dosagem , Óxidos de Nitrogênio/efeitos adversos , Óxidos de Nitrogênio/farmacocinética , Óxidos de Nitrogênio/farmacologia , Ratos , Ratos Wistar , Equivalência Terapêutica , Terapias em Estudo , Resultado do Tratamento
10.
Int J Toxicol ; 30(5): 466-77, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21868767

RESUMO

To support clinical development of S-nitrosoglutathione (GSNO) as a therapeutic agent, 28-day toxicology studies in rats and dogs were conducted. Rats (21-25/sex) and dogs (3-5/sex) were exposed for 4 hours or 1 hour, respectively, to inhaled GSNO (0, 3, 9.3, 19, and 28 mg/kg per d in rats and 0, 4.6, 9.0, and 16.2 mg/kg per d in dogs) or vehicle daily via a nebulizer. Animals were monitored throughout the 28-day dosing period and during a postexposure recovery period. Complete necropsy and tissue examinations were performed. Experimental end points included clinical pathology, toxicokinetics, and immunotoxicology. No biologically significant adverse findings were noted in either species, and the no observed adverse effect levels (NOAELs) under these conditions were the highest achieved doses (28 and 16.2 mg/kg per d in rats and dogs, respectively). These data demonstrate that GSNO is well tolerated in rodents and dogs and predict a favorable toxicity profile in humans, thus supporting future clinical development of GSNO or closely related compounds.


Assuntos
S-Nitrosoglutationa/farmacocinética , S-Nitrosoglutationa/toxicidade , Testes de Toxicidade/métodos , Administração por Inalação , Animais , Disponibilidade Biológica , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Determinação de Ponto Final , Eritrócitos , Feminino , Masculino , Nitratos/sangue , Nitratos/farmacocinética , Nitratos/urina , Nível de Efeito Adverso não Observado , Ratos , Ratos Wistar , Ovinos
11.
J Biomed Mater Res B Appl Biomater ; 93(2): 416-24, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20101734

RESUMO

Poly(vinyl alcohol) (PVA) is a biocompatible polymer already used in several pharmaceutical products. The purpose of this work was to investigate the influence of freezing-thawing cycles (F/T) on the in vitro diffusion and skin vasodilator properties of S-nitrosoglutathione (GSNO)-releasing PVA films. Films subjected to 1-, 3-, and 5-F/T showed an increase in crystallinity, which is associated with an increase in the radius of gyration of macropores from 155 to 180 nm. Diffusion coefficients (D) of GSNO decreased from 5.7 x 10(-7) to 2.0 x 10(-7) cm(2) s(-1) in 1 and 3 F/T films, respectively, and were inversely correlated with the increase in crystallinity, whereas 5-F/T films showed an anomalous increase in D (5.0 x 10(-7) cm(2) s(-1)). Topical release of GSNO from PVA films on the skin of healthy volunteers led to local vasodilation measured by laser Doppler flowmetry. A higher increase in local blood flow was observed for 5-F/T films reaching maximum tissue perfusion at 45 min with return toward basal level after 45 min, whereas 1-F/T films led to a lower increase in blood flow up to 98 min. These results show that F/T treatment can be used to modulate the diffusion properties and the topical vasodilator profile of GSNO-containing PVA films, what might allow the use of these materials as dermal wound dressings or for promoting local vasodilation in ischemic tissues.


Assuntos
Materiais Biocompatíveis , Sistemas de Liberação de Medicamentos , Membranas Artificiais , Álcool de Polivinil , S-Nitrosoglutationa/farmacocinética , Vasodilatadores/farmacocinética , Administração Tópica , Adulto , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Feminino , Humanos , Isquemia/tratamento farmacológico , Masculino , Porosidade , S-Nitrosoglutationa/administração & dosagem , Pele/irrigação sanguínea , Vasodilatação/efeitos dos fármacos , Vasodilatadores/administração & dosagem
12.
J Biol Chem ; 280(10): 8733-41, 2005 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-15611098

RESUMO

S-nitrosoglutathione (GSNO) denitrosation activity of recombinant human protein disulfide isomerase (PDI) has been kinetically characterized by monitoring the loss of the S-NO absorbance, using a NO electrode, and with the aid of the fluorogenic NOx probe 2,3-diaminonaphthalene. The initial rates of denitrosation as a function of [GSNO] displayed hyperbolic behavior irrespective of the method used to monitor denitrosation. The Km values estimated for GSNO were 65 +/- 5 microm and 40 +/- 10 microm for the loss in the S-NO bond and NO production (NO electrode or 2,3-diaminonaphthalene), respectively. Hemoglobin assay provided additional evidence that the final product of PDI-dependent GSNO denitrosation was NO*. A catalytic mechanism, involving a nitroxyl disulfide intermediate stabilized by imidazole (His160 a-domain or His589 a'-domain), which after undergoing a one-electron oxidation decomposes to yield NO plus dithiyl radical, has been proposed. Evidence for the formation of thiyl/dithiyl radicals during PDI-catalyzed denitrosation was obtained with 4-((9-acridinecarbonyl)-amino)-2,2,6,6-tetramethylpiperidine-1-oxyl. Evidence has also been obtained showing that in a NO- and O2-rich environment, PDI can form N2O3 in its hydrophobic domains. This "NO-charged PDI" can perform intra- and intermolecular S-nitrosation reactions similar to that proposed for serum albumin. Interestingly, reduced PDI was able to denitrosate S-nitrosated PDI (PDI-SNO) resulting in the release of NO. PDI-SNO, once formed, is stable at room temperature in the absence of reducing agent over the period of 2 h. It has been established that PDI is continuously secreted from cells that are net producers of NO-like endothelial cells. The present demonstration that PDI can be S-nitrosated and that PDI-SNO can be denitrosated by PDI suggests that this enzyme could be intimately involved in the transport of intracellular NO equivalents to the cell surface as well as the previous demonstration of PDI in the transfer of S-nitrosothiol-bound NO to the cytosol.


Assuntos
Isomerases de Dissulfetos de Proteínas/metabolismo , S-Nitrosoglutationa/farmacocinética , Clonagem Molecular , Eletroquímica , Escherichia coli/enzimologia , Hemoglobinas/metabolismo , Humanos , Cinética , Óxido Nítrico/análise , Óxido Nítrico/metabolismo , Proteínas Recombinantes/metabolismo
13.
Biochem J ; 387(Pt 2): 473-8, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15579132

RESUMO

The N-nitroso-derivative of melatonin, NOM (1-nitrosomelatonin), which has been demonstrated to be a NO* [oxidonitrogen*] donor in buffered solutions, is a new potential drug particularly in neurological diseases. The advantage of NOM, a very lipophilic drug, is its ability to release both melatonin and NO*, an easily diffusible free radical. In order to evaluate the distribution and the pharmacokinetics of NOM, [O-methyl-3H]NOM was administered to and followed in mice. A complementary method for monitoring NOM, EPR, was performed in vitro and ex vivo with (MGD)2-Fe2+ (iron-N-methyl-D-glucamine dithiocarbamate) complex as a spin trap. The behaviour of NOM was compared with that of GSNO (S-nitrosoglutathione), a hydrophilic NO* donor. In the first minutes following [O-methyl-3H]NOM intraperitoneal injection, the radioactivity was found in organs (6% in the liver, 1% in the kidney and 0.6% in the brain), but not in the blood. In both liver and brain, the radioactivity content decreased over time with similar kinetics reflecting the diffusion and metabolism of NOM and of its metabolites. Based on the characterization and the quantification of the EPR signal in vitro with NOM or GSNO using (MGD)2-Fe2+ complex in phosphate-buffered solutions, the detection of these nitroso compounds was realized ex vivo in mouse tissue extracts. (MGD)2-Fe2+-NO was observed in the brain of NOM-treated mice in the first 10 min following injection, revealing that NOM was able to cross the blood-brain barrier, while GSNO was not.


Assuntos
Melatonina/análogos & derivados , Compostos Nitrosos/farmacocinética , S-Nitrosoglutationa/farmacocinética , Animais , Barreira Hematoencefálica , Encéfalo/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Feminino , Fígado/metabolismo , Melatonina/farmacocinética , Camundongos , Camundongos Endogâmicos C57BL , Sorbitol/análogos & derivados , Marcadores de Spin , Tiocarbamatos , Distribuição Tecidual , Trítio
14.
Br J Pharmacol ; 141(7): 1204-13, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15006900

RESUMO

1. The involvement of peripheral nitric oxide (NO) in febrigenic signaling to the brain has been proposed because peripherally administered NO synthase (NOS) inhibitors attenuate lipopolysaccharide (LPS)-induced fever in rodents. However, how the unstable molecule of NO can reach the brain to trigger fever is unclear. It is also unclear whether NOS inhibitors attenuate fever by blocking febrigenic signaling or, alternatively, by suppressing thermogenesis in brown fat. 2. Male Wistar rats were chronically implanted with jugular catheters; their colonic and tail skin temperatures (T(c) and T(sk)) were monitored. 3. Study 1 was designed to determine whether the relatively stable, physiologically relevant forms of NO, that is, S-nitrosoalbumin (SNA) and S-nitrosoglutathione (SNG), are pyrogenic and whether they enhance LPS fever. At a neutral ambient temperature (T(a)) of 31 degrees C, afebrile or LPS (1 microg kg(-1), i.v.)-treated rats were infused i.v. with SNA (0.34 or 4.1 micromol kg(-1); the controls received NaNO(2) and albumin) or SNG (10 or 60 micromol kg(-1); the controls received glutathione). T(c) of SNA- or SNG-treated rats never exceeded that of the controls. 4. In Study 2, we tested whether the known fever-attenuating effect of the NOS inhibitor N(omega)-nitro-L-arginine methyl ester (L-NAME) at a subneutral T(a) (when fever is brought about by thermogenesis) also occurs at a neutral T(a) (when fever is brought about by skin vasoconstriction). At a subneutral T(a) of 24 degrees C, L-NAME (2.5 mg kg(-1), i.v.) attenuated LPS (10 microg kg(-1), i.v.) fever, presumably by inhibiting thermogenesis. At 31 degrees C, L-NAME enhanced LPS fever by augmenting skin vasoconstriction (T(sk) fall). 5. In summary, both SNA and SNG had no pyrogenic effect of their own and failed to enhance LPS fever; peripheral L-NAME attenuated only fever brought about by increased thermogenesis. It is concluded that NO is uninvolved in febrigenic signaling to the brain.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Encéfalo/fisiologia , Óxido Nítrico/fisiologia , Transdução de Sinais/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Regulação da Temperatura Corporal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Previsões , Glutationa/administração & dosagem , Glutationa/farmacocinética , Infusões Intravenosas , Isomerismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/efeitos adversos , Masculino , NG-Nitroarginina Metil Éster/administração & dosagem , NG-Nitroarginina Metil Éster/farmacocinética , Compostos Nitrosos , Ratos , Ratos Wistar , S-Nitrosoglutationa/administração & dosagem , S-Nitrosoglutationa/farmacocinética , Soroalbumina Bovina/administração & dosagem , Soroalbumina Bovina/farmacocinética , Transdução de Sinais/efeitos dos fármacos , Pele/irrigação sanguínea , Pele/efeitos dos fármacos , Temperatura Cutânea/efeitos dos fármacos , Temperatura Cutânea/fisiologia , Nitrito de Sódio/administração & dosagem , Nitrito de Sódio/farmacocinética , Vasoconstrição/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA