Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
ACS Macro Lett ; 13(3): 288-295, 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38368530

RESUMO

We report a facile stimuli-responsive strategy to generate reactive oxygen and nitrogen species (ROS and RNS) in the biological milieu from a photocleavable water-soluble block copolymer under visible light irradiation (427 nm, 2.25 mW/cm2). An anthraquinone-based water-soluble polymeric nitric oxide (NO) donor (BCPx-NO) is synthesized, which exhibits NO release in the range of 40-65 µM within 10 h of photoirradiation with a half-life of 30-103 min. Additionally, BCPx-NO produces peroxynitrite (ONOO-) and singlet oxygen (1O2) under photoirradiation. To understand the mechanism of NO release and photolysis of the functional group under blue light, we prepared a small-molecule anthraquinone-based N-nitrosamine (NOD). The cellular investigation of the effect of spatiotemporally controlled ONOO- and 1O2 generation from the NO donor polymeric nanoparticles in a triple negative breast adenocarcinoma (MDA-MB-231) under visible light irradiation (white light, 5.83 mW/cm2; total dose 31.5 J/cm2) showed an IC50 of 0.6 mg/mL. The stimuli-responsive strategy using a photolabile water-soluble block copolymer employed to generate ROS and RNS in a biological setting widens the horizon for their potential in cancer therapy.


Assuntos
Neoplasias , Ácido Peroxinitroso , Humanos , Ácido Peroxinitroso/uso terapêutico , Espécies Reativas de Oxigênio/uso terapêutico , Polímeros/uso terapêutico , Espécies Reativas de Nitrogênio/uso terapêutico , Luz , Oxigênio/uso terapêutico , Óxido Nítrico/uso terapêutico , Antraquinonas/uso terapêutico , Neoplasias/tratamento farmacológico
2.
J Adv Res ; 56: 43-56, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36958586

RESUMO

INTRODUCTION: Chemodynamic therapy (CDT) holds great promise in achieving cancer therapy through Fenton and Fenton-like reactions, which generate highly toxic reactive species. However, CDT is limited by the lower amount of catalyst ions that can decompose already existing intracellular H2O2 and produce reactive oxygen species (ROS) to attain a therapeutic outcome. OBJECTIVES: To overcome these limitations, a tailored approach, which utilizes dual metals cations (Ag+, Fe2+) based silver pentacyanonitrosylferrate or silver nitroprusside (AgNP) were developed for Fenton like reactions that can specifically kill cancer cells by taking advantage of tumor acidic environment without used of any external stimuli. METHODS: A simple solution mixing procedure was used to synthesize AgNP as CDT agent. AgNP were structurally and morphologically characterized, and it was observed that a minimal dose of AgNP is required to destroy cancer cells with limited effects on normal cells. Moreover, comprehensive in vitro studies were conducted to evaluate antitumoral mechanism. RESULTS: AgNP have an effective ability to decompose endogenous H2O2 in cells. The decomposed endogenous H2O2 generates several different types of reactive species (•OH, O2•-) including peroxynitrite (ONOO-) species as apoptotic inducers that kill cancer cells, specifically. Cellular internalization data demonstrated that in short time, AgNP enters in lysosomes, avoid degradation and due to the acidic pH of lysosomes significantly generate high ROS levels. These data are further confirmed by the activation of different oxidative genes. Additionally, we demonstrated the biocompatibility of AgNP on mouse liver and ovarian organoids as an ex vivo model while AgNP showed the therapeutic efficacy on patient derived tumor organoids (PDTO). CONCLUSION: This work demonstrates the therapeutic application of silver nitroprusside as a multiple ROS generator utilizing Fenton like reaction. Thereby, our study exhibits a potential application of CDT against HGSOC (High Grade Serous Ovarian Cancer), a deadly cancer through altering the redox homeostasis.


Assuntos
Neoplasias , Prata , Camundongos , Animais , Humanos , Espécies Reativas de Oxigênio/metabolismo , Prata/química , Prata/farmacologia , Prata/uso terapêutico , Nitroprussiato/farmacologia , Nitroprussiato/uso terapêutico , Ácido Peroxinitroso/uso terapêutico , Peróxido de Hidrogênio/química , Neoplasias/tratamento farmacológico
3.
Anal Biochem ; 682: 115338, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37802174

RESUMO

Osteoarthritis (OA) is the most common type of joint disease, which is difficult to treat, but early standardized diagnosis and treatment can effectively alleviate the pain and symptoms of patients. Therefore, it is important to construct an effective tool to assist in the early diagnosis and evaluation of the therapeutic effect of OA. In this work, a near-infrared (NIR) fluorescence-activated fluorescent probe, YB-1, was constructed for the evaluation of the diagnostic and therapeutic efficacy of OA via detection and imaging of the biomarker of ONOO- in inflammatory cells and mice osteoarthritis models. YB-1 exhibited high selectivity, high sensitivity, and a high ratio yield (I668/I0) fluorescence increasing (∼30 folds). Besides, YB-1 can be used effectively to image endogenous and exogenous ONOO- in living human chondrocytes cells (TC28a2), as well as to evaluate the effect of drug (Chrysosplenol D, CD) treatment in IL-1ß-induced inflammatory cells model. Interestingly, YB-1 was available for OONO- imaging analysis in the collagenase-induced mice OA models and assessment of the effect of CD treatment in mice OA models, with good results. Thus, the newly constructed YB-1 is a powerful molecular tool for the diagnosis and treatment of OA-related diseases.


Assuntos
Corantes Fluorescentes , Osteoartrite , Camundongos , Animais , Humanos , Corantes Fluorescentes/farmacologia , Ácido Peroxinitroso/farmacologia , Ácido Peroxinitroso/uso terapêutico , Osteoartrite/diagnóstico por imagem , Condrócitos , Diagnóstico por Imagem , Modelos Animais de Doenças
4.
Cell Rep Med ; 3(10): 100787, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36260983

RESUMO

A study by Tcyganov et al.1 demonstrates that peroxynitrite, an oxidant abundant in the tumor microenvironment, changes the repertoire of MHC class I peptides presented by tumors and limits immune recognition. Peroxynitrite inhibition in combination with immune checkpoint blockade enhances efficacy preclinically.


Assuntos
Evasão da Resposta Imune , Neoplasias , Humanos , Ácido Peroxinitroso/uso terapêutico , Inibidores de Checkpoint Imunológico , Antígenos de Neoplasias/uso terapêutico , Antígenos de Histocompatibilidade Classe I , Neoplasias/tratamento farmacológico , Peptídeos/uso terapêutico , Oxidantes/uso terapêutico , Microambiente Tumoral
5.
Angew Chem Int Ed Engl ; 61(40): e202209793, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-35916871

RESUMO

Herein, we report an activatable near-infrared (NIR) afterglow theranostic prodrug that circumvents high background noise interference caused by external light excitation. The prodrug can release hydroxycamptothecin (HCPT) in response to the high intratumoral peroxynitrite level associated with immunogenic cell death (ICD), and synchronously activate afterglow signal to monitor the drug release process and cold-to-hot tumor transformation. The prodrug itself is an ICD inducer achieved by photodynamic therapy (PDT). PDT initiates ICD and recruits first-arrived neutrophils to secrete peroxynitrite to trigger HCPT release. Intriguingly, we demonstrate that HCPT can significantly amplify PDT-mediated ICD process. The prodrug thus shows a self-sustainable ICD magnification effect by establishing an "ICD-HCPT release-amplified ICD" cycling loop. In vivo studies demonstrate that the prodrug can eradicate existing tumors and prevent further tumor recurrence through antitumor immune response.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Pró-Fármacos , Linhagem Celular Tumoral , Humanos , Morte Celular Imunogênica , Neoplasias/tratamento farmacológico , Ácido Peroxinitroso/uso terapêutico , Medicina de Precisão , Pró-Fármacos/metabolismo
6.
Sheng Li Xue Bao ; 74(1): 93-109, 2022 Feb 25.
Artigo em Chinês | MEDLINE | ID: mdl-35199130

RESUMO

Diabetic nephropathy is a microvascular complication of diabetes. Its etiology involves metabolic disorder-induced endothelial dysfunction. Endothelium-derived nitric oxide (NO) plays an important role in a number of physiological processes, including glomerular filtration and endothelial protection. NO dysregulation is an important pathogenic basis of diabetic nephropathy. Hyperglycemia and dyslipidemia can lead to oxidative stress, chronic inflammation and insulin resistance, thus affecting NO homeostasis regulated by endothelial nitric oxide synthase (eNOS) and a conglomerate of related proteins and factors. The reaction of NO and superoxide (O2.-) to form peroxynitrite (ONOO-) is the most important pathological NO pathway in diabetic nephropathy. ONOO- is a hyper-reactive oxidant and nitrating agent in vivo which can cause the uncoupling of eNOS. The uncoupled eNOS does not produce NO but produces superoxide. Thus, eNOS uncoupling is a critical contributor of NO dysregulation. Understanding the regulatory mechanism of NO and the effects of various pathological conditions on it could reveal the pathophysiology of diabetic nephropathy, potential drug targets and mechanisms of action. We believe that increasing the stability and activity of eNOS dimers, promoting NO synthesis and increasing NO/ONOO- ratio could guide the development of drugs to treat diabetic nephropathy. We will illustrate these actions with some clinically used drugs as examples in the present review.


Assuntos
Diabetes Mellitus , Nefropatias Diabéticas , Nefropatias Diabéticas/tratamento farmacológico , Endotélio Vascular , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo III/farmacologia , Óxido Nítrico Sintase Tipo III/uso terapêutico , Estresse Oxidativo , Ácido Peroxinitroso/metabolismo , Ácido Peroxinitroso/farmacologia , Ácido Peroxinitroso/uso terapêutico
7.
Oncol Rep ; 45(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33760187

RESUMO

Tumors of the abdominal cavity, such as colorectal, pancreatic and ovarian cancer, frequently metastasize into the peritoneum. Large numbers of metastatic nodules hinder curative surgical resection, necessitating lavage with hyperthermic intraperitoneal chemotherapy (HIPEC). However, HIPEC not only causes severe side effects but also has limited therapeutic efficacy in various instances. At the same time, the age of immunotherapies such as biological agents, checkpoint­ inhibitors or immune­cell therapies, increasingly emphasizes the critical role of anticancer immunity in targeting malignancies. The present study investigated the ability of three types of long­lived reactive species (oxidants) to inactivate cancer cells and potentially complement current HIPEC regimens, as well as to increase tumor cell expression of danger signals that stimulate innate immunity. The human abdominal cancer cell lines HT­29, Panc­01 and SK­OV­3 were exposed to different concentrations of hydrogen peroxide (H2O2), hypochlorous acid (HOCl) and peroxynitrite (ONOO­). Metabolic activity was measured, as well as determination of cell death and danger signal expression levels via flow cytometry and detection of intracellular oxidation via high­content microscopy. Oxidation of tumor decreased intracellular levels of the antioxidant glutathione and induced oxidation in mitochondria, accompanied by a decrease in metabolic activity and an increase in regulated cell death. At similar concentrations, HOCl showed the most potent effects. Non­malignant HaCaT keratinocytes were less affected, suggesting the approach to be selective to some extent. Pro­immunogenic danger molecules were investigated by assessing the expression levels of calreticulin (CRT), and heat­shock protein (HSP)70 and HSP90. CRT expression was greatest following HOCl and ONOO­ treatment, whereas HOCl and H2O2 resulted in the greatest increase in HSP70 and HSP90 expression levels. These results suggested that HOCl may be a promising agent to complement current HIPEC regimens targeting peritoneal carcinomatosis.


Assuntos
Alarminas/metabolismo , Ácido Hipocloroso/farmacologia , Neoplasias Peritoneais/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Feminino , Células HaCaT , Humanos , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/uso terapêutico , Quimioterapia Intraperitoneal Hipertérmica/métodos , Ácido Hipocloroso/uso terapêutico , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Oxirredução/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Neoplasias Peritoneais/secundário , Peritônio/efeitos dos fármacos , Peritônio/patologia , Ácido Peroxinitroso/farmacologia , Ácido Peroxinitroso/uso terapêutico
8.
Angew Chem Int Ed Engl ; 60(9): 4720-4731, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33210779

RESUMO

Photoacoustic (PA) technology can transform light energy into acoustic wave, which can be used for either imaging or therapy that depends on the power density of pulsed laser. Here, we report photosensitizer-free polymeric nanocapsules loaded with nitric oxide (NO) donors, namely NO-NCPs, formulated from NIR light-absorbable amphiphilic polymers and a NO-releasing donor, DETA NONOate. Controlled NO release and nanocapsule dissociation are achieved in acidic lysosomes of cancer cells. More importantly, upon pulsed laser irradiation, the PA cavitation can excite water to generate significant reactive oxygen species (ROS) such as superoxide radical (O2.- ), which further spontaneously reacts with the in situ released NO to burst highly cytotoxic peroxynitrite (ONOO- ) in cancer cells. The resultant ONOO- generation greatly promotes mitochondrial damage and DNA fragmentation to initiate programmed cancer cell death. Apart from PA imaging, PA cavitation can intrinsically amplify reactive species via photosensitization-free materials for promising disease theranostics.


Assuntos
Raios Infravermelhos , Nanocápsulas/química , Ácido Peroxinitroso/química , Polímeros/química , Espécies Reativas de Oxigênio/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Clorofilídeos , Dano ao DNA/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/química , Ácido Peroxinitroso/uso terapêutico , Ácido Peroxinitroso/toxicidade , Técnicas Fotoacústicas , Porfirinas/farmacologia , Porfirinas/uso terapêutico , Superóxidos/metabolismo , Nanomedicina Teranóstica , Transplante Homólogo
9.
CNS Neurosci Ther ; 21(7): 585-90, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25996167

RESUMO

AIM: Hemorrhagic transformation (HT) is a major complication of delayed tissue plasminogen activator (t-PA) treatment in ischemic stroke. We aimed to explore whether peroxynitrite decomposition catalyst (PDC) could prevent such complication. METHODS: Male Sprague-Dawley (SD) rats were subjected to middle cerebral artery occlusion (MCAO) with t-PA (10 mg/kg) or t-PA plus FeTMPyP (3 mg/kg, a representative PDC) at MCAO for 2 or 5 h and reperfusion for 22 or 19 h, respectively. HT was assessed with hemoglobin assay. Neurological deficit was evaluated with Modified Neurological Severity Score (mNSS). Peroxynitrite formation was examined by detecting 3-nitrotyrosine (3-NT) formation. The expression and activity of MMP-9/MMP-2 were assessed by Western blotting and gelatin zymography. RESULTS: t-PA treatment at 2 h of MCAO did not induce HT but attenuated neurological deficit, whereas treatment at 5 h significantly induced HT and worsened the neurological outcome. Such complications were prevented by FeTMPyP cotreatment. Early t-PA treatment inhibited 3-NT and MMP-9/MMP-2 expression, whereas delayed treatment induced 3-NT and MMP-9/MMP-2 expression and activity. FeTMPyP cotreatment downregulated 3-NT and inhibited MMP-9/MMP-2 in both time points. CONCLUSION: Peroxynitrite decomposition catalyst could prevent hemorrhagic transformation and improve neurological outcome ischemic rat brains with delayed t-PA treatment via inhibiting peroxynitrite-mediated MMP activation.


Assuntos
Fibrinolíticos/efeitos adversos , Hemorragia/induzido quimicamente , Hemorragia/tratamento farmacológico , Hemostáticos/uso terapêutico , Ácido Peroxinitroso/uso terapêutico , Ativador de Plasminogênio Tecidual/efeitos adversos , Animais , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Infarto da Artéria Cerebral Média/tratamento farmacológico , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Doenças do Sistema Nervoso/etiologia , Ratos , Ratos Sprague-Dawley , Reperfusão , Índice de Gravidade de Doença
10.
Transplant Proc ; 41(9): 3603-10, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19917352

RESUMO

BACKGROUND: In an acute kidney transplant rejection rat model, we demonstrated that manganese superoxide dismutase (MnSOD) activity was significantly reduced and MnSOD was nitrated by peroxynitrite (ONOO(-)), resulting in tissue injury. We examined whether tissue injury was reduced after external supplementation of recombinant human MnSOD in a rat renal ischemia-reperfusion injury model. METHODS: Male Brown-Norway rats underwent dissection of the right kidney. The animals were divided into 3 groups. The controls had the left renal blood vessels clamped for 90 minutes to induce ischemia, followed by reperfusion for 16 hours. In the intraperitoneal administration group, MnSOD was administered 30 minutes before ischemia and immediately before reperfusion. In the sham group, neither ischemia nor reperfusion was performed. After reperfusion, blood was collected, the left kidney was dissected and renal function and tissue injury were evaluated. RESULTS: Serum creatinine and K(+), blood urea nitrogen, and aspartate aminotransferase activity decreased significantly, whereas serum Na(+) and renal function improved in the MnSOD group compared with the control and sham groups. On hematoxylin and eosin staining, the histological score indicated that acute tubular necrosis was significantly reduced by MnSOD administration. Periodic acid-Schiff staining was absent in the nonadministration group, whereas it persisted in the MnSOD group. In the proximal renal tubules a large proportion of anti-nitrotyrosine staining was present before but absent after MnSOD administration. CONCLUSIONS: MnSOD administration improved renal function and reduced tissue injury. It may also reduce tissue injury in acute kidney transplant rejection and other tissue injuries caused by similar molecular mechanisms.


Assuntos
Rim/metabolismo , Ácido Peroxinitroso/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Superóxido Dismutase/uso terapêutico , Animais , Nitrogênio da Ureia Sanguínea , Clonagem Molecular , Primers do DNA , Modelos Animais de Doenças , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Nefrectomia , Ácido Peroxinitroso/uso terapêutico , Reação em Cadeia da Polimerase , Potássio/sangue , Ratos , Ratos Endogâmicos BN , Proteínas Recombinantes/uso terapêutico , Superóxido Dismutase/genética
12.
J Pharmacol Exp Ther ; 328(3): 777-84, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19033557

RESUMO

Oxidative/nitrative stress caused by peroxynitrite, the reaction product of superoxide (O2(.-)) and nitric oxide (NO), is the primary cause of myocardial ischemia/reperfusion injury. The present study determined whether INO-4885 [5,10,15,20-tetra[N-(benzyl-4'-carboxylate)-2-pyridinium]-21H,23H-porphine iron(III) chloride], a new peroxynitrite decomposition catalyst, may provide cellular protection and protect heart from myocardial ischemia/reperfusion injury. Adult male mice were subjected to 30 min of ischemia and 3 or 24 h of reperfusion. Mice were randomized to receive vehicle, INO-4885 without catalytic moiety, or INO-4885 (3-300 microg/kg i.p.) 10 min before reperfusion. Infarct size, apoptosis, nitrotyrosine content, NO/O2(.-) production, and inducible nitric-oxide synthase (iNOS)/NADPH oxidase expression were determined. INO-4885 treatment reduced ischemia/reperfusion-induced protein nitration and caspase 3 activation in a dose-dependent fashion in the range of 3 to 100 microg/kg. However, doses exceeding 100 microg/kg produced nonspecific effects and attenuated its protective ability. At the optimal dose (30 microg/kg), INO-4885 significantly reduced infarct size (p < 0.01), decreased apoptosis (p < 0.01), and reduced tissue nitrotyrosine content (p < 0.01). As expected, INO-4885 had no effect on ischemia/reperfusion-induced iNOS expression and NO overproduction. To our surprise, this compound significantly reduced superoxide production and partially blocked NADPH oxidase overexpression in the ischemic/reperfused cardiac tissue. Additional experiments demonstrated that INO-4885 provided better cardioprotection than N-(3-(aminomethyl)benzyl)acetamidine (1400W, a selective iNOS inhibitor), apocynin (an NADPH oxidase inhibitor), or Tiron (a cell-permeable superoxide scavenger). Taken together, our data demonstrated that INO-4885 is a cardioprotective molecule that attenuates myocardial reperfusion injury by facilitating peroxynitrite decomposition and inhibiting NADPH oxidase-derived O2(.-) production.


Assuntos
Coração/fisiopatologia , Metaloporfirinas/uso terapêutico , Infarto do Miocárdio/prevenção & controle , Ácido Peroxinitroso/uso terapêutico , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Coração/efeitos dos fármacos , Coração/fisiologia , Masculino , Camundongos , NADPH Oxidases/efeitos dos fármacos , NADPH Oxidases/genética , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
13.
Proc Natl Acad Sci U S A ; 105(40): 15511-6, 2008 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-18829442

RESUMO

CNS tissues are protected from circulating cells and factors by the blood-brain barrier (BBB), a specialization of the neurovasculature. Outcomes of the loss of BBB integrity and cell infiltration into CNS tissues can differ vastly. For example, elevated BBB permeability is closely associated with the development of neurological disease in experimental allergic encephalomyelitis (EAE) but not during clearance of the attenuated rabies virus CVS-F3 from the CNS tissues. To probe whether differences in the nature of BBB permeability changes may contribute to the pathogenesis of acute neuroinflammatory disease, we compared the characteristics of BBB permeability changes in mice with EAE and in mice clearing CVS-F3. BBB permeability changes are largely restricted to the cerebellum and spinal cord in both models but differ in the extent of leakage of markers of different size and in the nature of cell accumulation in the CNS tissues. The accumulation in the CNS tissues of CD4 T cells expressing mRNAs specific for IFN-gamma and IL-17 is common to both, but iNOS-positive cells invade into the CNS parenchyma only in EAE. Mice that have been immunized with myelin basic protein (MBP) and infected exhibit the features of EAE. Treatment with the peroxynitrite-dependent radical scavenger urate inhibits the invasion of iNOS-positive cells into the CNS tissues and the development of clinical signs of EAE without preventing the loss of BBB integrity in immunized/infected animals. These findings indicate that BBB permeability changes can occur in the absence of neuropathology provided that cell invasion is restricted.


Assuntos
Autoimunidade , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/virologia , Sistema Nervoso Central/imunologia , Encefalomielite Autoimune Experimental/imunologia , Raiva/imunologia , Animais , Barreira Hematoencefálica/patologia , Movimento Celular , Cerebelo/metabolismo , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Feminino , Sequestradores de Radicais Livres/metabolismo , Imuno-Histoquímica , Interferon gama/imunologia , Interleucina-17/imunologia , Camundongos , Camundongos Endogâmicos , Óxido Nítrico Sintase/metabolismo , Ácido Peroxinitroso/metabolismo , Ácido Peroxinitroso/uso terapêutico , Raiva/metabolismo , Vírus da Raiva/imunologia , Vírus da Raiva/metabolismo
14.
Diabetes ; 57(4): 889-98, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18285558

RESUMO

OBJECTIVE: Recently we have shown that diabetes-induced retinal neurodegeneration positively correlates with oxidative stress and peroxynitrite. Studies also show that peroxynitrite impairs nerve growth factor (NGF) survival signaling in sensory neurons. However, the causal role of peroxynitrite and the impact of tyrosine nitration on diabetes-induced retinal neurodegeneration and NGF survival signaling have not been elucidated. RESEARCH DESIGN AND METHODS: Expression of NGF and its receptors was examined in retinas from human and streptozotocin-induced diabetic rats and retinal ganglion cells (RGCs). Diabetic animals were treated with FeTPPS (15 mg x kg(-1) x day(-1) ip), which catalytically decomposes peroxynitrite to nitrate. After 4 weeks of diabetes, retinal cell death was determined by TUNEL assay. Lipid peroxidation and nitrotyrosine were determined using MDA assay, immunofluorescence, and Slot-Blot analysis. Expression of NGF and its receptors was determined by enzyme-linked immunosorbent assay (ELISA), real-time PCR, immunoprecipitation, and Western blot analyses. RESULTS: Analyses of retinal neuronal death and NGF showed ninefold and twofold increases, respectively, in diabetic retinas compared with controls. Diabetes also induced increases in lipid peroxidation, nitrotyrosine, and the pro-apoptotic p75(NTR) receptor in human and rat retinas. These effects were associated with tyrosine nitration of the pro-survival TrkA receptor, resulting in diminished phosphorylation of TrkA and its downstream target, Akt. Furthermore, peroxynitrite induced neuronal death, TrkA nitration, and activation of p38 mitogen-activated protein kinase (MAPK) in RGCs, even in the presence of exogenous NGF. FeTPPS prevented tyrosine nitration, restored NGF survival signal, and prevented neuronal death in vitro and in vivo. CONCLUSIONS: Together, these data suggest that diabetes-induced peroxynitrite impairs NGF neuronal survival by nitrating TrkA receptor and enhancing p75(NTR) expression.


Assuntos
Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus/fisiopatologia , Neuropatias Diabéticas/tratamento farmacológico , Degeneração Neural/prevenção & controle , Fator de Crescimento Neural/genética , Nervo Óptico/fisiopatologia , Ácido Peroxinitroso/uso terapêutico , Animais , Cadáver , Morte Celular , Diabetes Mellitus/patologia , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Pessoa de Meia-Idade , Fator de Crescimento Neural/antagonistas & inibidores , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/patologia , RNA Mensageiro/genética , Ratos , Valores de Referência , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Estreptozocina , Tirosina/análogos & derivados , Tirosina/análise
15.
Transplantation ; 80(10): 1455-60, 2005 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-16340791

RESUMO

BACKGROUND: Peroxynitrite anions may play a role in normothermic renal ischemia and reperfusion. The purpose of this study was to determine if endogenous peroxynitrite anion is involved in renal preservation injury. METHODS: Experiments were conducted in isolated canine renal tubules and in a canine autotransplant model of hypothermic preservation injury. RESULTS: Isolated renal tubules demonstrated progressive loss of membrane transport function after reperfusion with increasing cold storage times in UW solution as assessed by tetraethylammonium cation transport (TEA). This transport defect was not altered by reperfusion in the presence of WW85, a peroxynitrite decomposition catalyst. Likewise, tubule LDH release was not altered by WW85. Renal tubules did not demonstrate any evidence of peroxynitrite formation after cold storage (0-120 h) or after subsequent reperfusion in vitro as measured by nitrotyrosine adduct formation. Addition of exogenous peroxynitrite (1 mM) directly to freshly isolated renal tubules produced strong nitrotyrosine signals but failed to alter membrane function (TEA uptake). Conversely, SIN-1, a peroxynitrite generator molecule, failed to produce a nitrotyrosine signal in extracted renal tubule proteins but significantly impaired transport function. Finally, function of cold stored canine autografts was not affected by the scavenging of peroxynitrite anions (WW85) before kidney harvest and immediately at reperfusion. Tissue biopsies from cold stored kidney autografts also failed to show evidence of peroxynitrite synthesis either after cold storage (72 h) or after kidney transplantation (60 min reperfusion). CONCLUSIONS: This study concludes that peroxynitrite anions are not formed and are not involved in renal preservation injury.


Assuntos
Criopreservação , Rim , Preservação de Órgãos/efeitos adversos , Ácido Peroxinitroso/biossíntese , Traumatismo por Reperfusão/fisiopatologia , Animais , Creatinina/sangue , Cães , Rim/efeitos dos fármacos , Rim/fisiopatologia , Transplante de Rim , Túbulos Renais/enzimologia , L-Lactato Desidrogenase/metabolismo , Ácido Peroxinitroso/uso terapêutico , Ácido Peroxinitroso/toxicidade
16.
Pharmacol Res ; 49(1): 7-16, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14597146

RESUMO

The aims of the present study were to investigate the effects of exogenous peroxynitrite (ONOO(-)) on reperfusion arrhythmias in anaesthetized rats, and to detect endogenous and exogenous ONOO(-)-induced chemiluminescence (CL) signals in rat whole blood, which was collected during baseline and in the first minute of reperfusion. ONOO(-) infusion in ischemia/reperfusion (I/R) applied groups caused significant decreases in mean arterial pressure (MAP) and heart rate (HR). Ventricular fibrillation (VF) incidences in the vehicle, ONOO(-), and dec-ONOO(-) infused groups were 63, 100, and 20%, respectively. In control group CL signal was 136+/-34mV during the resting period and was increased to 336+/-57mV with reperfusion. Also, the effects of SOD+CAT, L-NAME and urate were investigated. Ventricular tachycardia (VT) incidence was decreased significantly in SOD+CAT and urate; VF incidence was decreased significantly in SOD+CAT applied groups. CL signals were inhibited with SOD+CAT, L-NAME, and urate. Exogenous ONOO(-) infusion during I/R was also investigated. CL signal in exogenously ONOO(-) infused group is increased 423% during reperfusion. Only urate infused group VF incidence was decreased significantly. CL signals of ONOO(-) infused groups were inhibited by SOD+CAT, L-NAME, and urate. Based on the results of the current study, ONOO(-) seems to be one of the key mediators of reperfusion arrhythmias in anaesthetized rats.


Assuntos
Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Ácido Peroxinitroso/uso terapêutico , Animais , Pressão Sanguínea/efeitos dos fármacos , Catalase/farmacologia , Catalase/uso terapêutico , Esquema de Medicação , Quimioterapia Combinada , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Infusões Intravenosas , Medições Luminescentes , Masculino , Traumatismo por Reperfusão Miocárdica/complicações , Traumatismo por Reperfusão Miocárdica/mortalidade , N-Formilmetionina Leucil-Fenilalanina/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Ácido Peroxinitroso/sangue , Ácido Peroxinitroso/química , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/farmacologia , Superóxido Dismutase/uso terapêutico , Taquicardia Ventricular/complicações , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/mortalidade , Ácido Úrico/farmacologia , Ácido Úrico/uso terapêutico , Fibrilação Ventricular/complicações , Fibrilação Ventricular/tratamento farmacológico , Fibrilação Ventricular/mortalidade , Visão Ocular/efeitos dos fármacos
17.
Ann Neurol ; 54(4): 451-8, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14520656

RESUMO

Hearing loss is the most frequent long-term complication of pneumococcal meningitis, affecting up to 40% of survivors. Unfortunately, adjuvant therapy with dexamethasone has failed to satisfactorily reduce its incidence. Therefore, we evaluated the use of antioxidants for the adjunctive therapy of meningitis-associated deafness. Eighteen hours after intracisternal injection of 7.5 x 10(5) colony-forming units of Streptococcus pneumoniae, rats were treated systemically either with ceftriaxone and the antioxidants and peroxynitrite scavengers Mn(III)tetrakis(4-benzoic acid)-porphyrin (MnTBAP) or N-acetyl-L-cysteine (NAC) or placebo (1 ml phosphate-buffered saline) for 4 days. Hearing was assessed by auditory brainstem response audiometry. Adjunctive antioxidant therapy significantly reduced the long-term hearing loss (14 days after infection) for square wave impulses (mean hearing loss +/- SD: ceftriaxone and placebo, 45+/-26 dB; ceftriaxone and MnTBAP, 9+/-23 dB; ceftriaxone and NAC, 19+/-30 dB) as well as 1 kHz (ceftriaxone and placebo, 28+/-19 dB; ceftriaxone and MnTBAP, 10+/-16 dB; ceftriaxone and NAC, 10+/-17 dB), and 10 kHz tone bursts (ceftriaxone and placebo, 62+/-27 dB; ceftriaxone and MnTBAP, 16+/-13 dB; ceftriaxone and NAC, 25+/-26 dB). Furthermore, both antioxidants attenuated the morphological correlates of meningogenic hearing loss, namely, long-term blood-labyrinth barrier disruption, spiral ganglion neuronal loss, and fibrous obliteration of the perilymphatic spaces. Adjuvant antioxidant therapy is highly otoprotective in meningitis and therefore is a promising future treatment option.


Assuntos
Antioxidantes/uso terapêutico , Perda Auditiva/prevenção & controle , Meningite Pneumocócica/complicações , Ácido Peroxinitroso/uso terapêutico , Acetilcisteína/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Audiometria , Ceftriaxona/uso terapêutico , Contagem de Células/métodos , Cóclea/efeitos dos fármacos , Cóclea/patologia , Modelos Animais de Doenças , Interações Medicamentosas , Azul Evans/metabolismo , Potenciais Evocados Auditivos do Tronco Encefálico/fisiologia , Perda Auditiva/etiologia , Labirintite/microbiologia , Labirintite/patologia , Masculino , Meningite Pneumocócica/microbiologia , Metaloporfirinas/uso terapêutico , Ratos , Ratos Wistar , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/patologia , Fatores de Tempo
18.
Eur J Pharmacol ; 449(1-2): 177-81, 2002 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-12163122

RESUMO

We evaluated the effect of different peroxynitrite scavengers for adjunctive therapy of experimental bacterial meningitis. Twenty hours after intracisternal injection of Streptococcus pneumoniae, rats were treated with ceftriaxone [100 mg/kg intraperitoneal (i.p.)] and either urate (300 mg/kg i.p.), Mn(III)tetrakis(4-benzoic acid)porphyrin (MnTBAP, 15 mg/kg i.p.), ascorbate (100 mg/kg i.p.), or urate (300 mg/kg i.p.) + ascorbate (100 mg/kg i.p.). Six hours after initiation of treatment, the cerebrospinal fluid (CSF) pleocytosis was significantly (p<0.05) reduced by urate (8697 +/- 1526 cells/microl) and MnTBAP (8542 +/- 4059 cells/microl) vs. ceftriaxone alone (15,793 +/- 3202 cells/microl). Brain concentrations of proinflammatory cytokines [interleukin-1beta (IL-beta), interleukin-6 (IL-6), and macrophage inflammatory protein-2 (MIP-2)] were also reduced by urate and MnTBAP. The intracranial hypertension was significantly reduced by MnTBAP (14.0 +/- 5.4 mm Hg), but not by urate (25.5 +/- 7.1 mm Hg) vs. ceftriaxone alone (22.5 +/- 5.9 mm Hg). Ascorbate alone had no effect on CSF pleocytosis (15,775 +/- 7058 cells/microl), intracranial pressure (25.6 +/- 8.8 mm Hg), and brain cytokine concentrations. However, the combination of urate and ascorbate was as effective as MnTBAP (CSF pleocytosis: 5392 +/- 4232 cells/microl, intracranial pressure: 13.3 +/- 6.9 mm Hg).


Assuntos
Sequestradores de Radicais Livres/uso terapêutico , Meningite Pneumocócica/tratamento farmacológico , Ácido Peroxinitroso/uso terapêutico , Animais , Química Encefálica/efeitos dos fármacos , Química Encefálica/fisiologia , Ceftriaxona/uso terapêutico , Cefalosporinas/uso terapêutico , Quimiocina CXCL2 , Quimiocinas/metabolismo , Citocinas/metabolismo , Indicadores e Reagentes , Interleucina-1/metabolismo , Interleucina-6/metabolismo , Masculino , Meningite Pneumocócica/líquido cefalorraquidiano , Meningite Pneumocócica/microbiologia , Monocinas/metabolismo , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA