Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Hepatol ; 79(4): 977-988, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37482222

RESUMEN

BACKGROUND & AIMS: ß-blockers reduce hepatic venous pressure gradient (HVPG) by decreasing portal inflow, with no reduction in intrahepatic vascular resistance. 5-Methyltetrahydrofolate (5-MTHF) can prevent oxidative loss of tetrahydrobiopterin (BH4), a cofactor for endothelial nitric oxide synthase coupling. It also converts homocysteine (tHcy) into methionine and enables the degradation of asymmetric dimethylarginine (ADMA), an inhibitor of endothelial nitric oxide synthase. The aim of this study was to evaluate the effects of 5-MTHF in combination with propranolol on HVPG and nitric oxide bioavailability markers in patients with cirrhosis and portal hypertension. METHOD: Sixty patients with cirrhosis and HVPG ≥12 mmHg were randomized 1:1 to receive treatment with 5-MTHF+propranolol or placebo+propranolol for 90 days under double-blind conditions. HVPG and markers of nitric oxide bioavailability (BH4, ADMA and tHcy) were measured again at the end of treatment. RESULTS: Groups were similar in terms of baseline clinical and hemodynamic data and nitric oxide bioavailability markers. HVPG decreased in both groups, but the magnitude of the change was significantly greater in the group treated with 5-MTHF+propranolol compared to placebo+propranolol (percentage decrease, 20 [29-9] vs. 12.5 [22-0], p = 0.028), without differences in hepatic blood flow. At the end of treatment, 5-MTHF+propranolol (vs. placebo+propranolol) was associated with higher BH4 (1,101.4 ± 1,413.3 vs. 517.1 ± 242.8 pg/ml, p <0.001), lower ADMA (109.3 ± 52.7 vs. 139.9 ± 46.7 µmol/L, p = 0.027) and lower tHcy (µmol/L, 11.0 ± 4.6 vs. 15.4 ± 7.2 µmol/L, p = 0.010) plasma levels. CONCLUSION: In patients with cirrhosis and portal hypertension, 5-MTHF administration significantly enhanced the HVPG reduction achieved with propranolol. This effect appears to be mediated by improved nitric oxide bioavailability in the hepatic microcirculation. CLINICAL TRIAL EUDRACT NUMBER: 2014-002018-21. IMPACT AND IMPLICATIONS: Currently, the pharmacological prevention of cirrhosis complications due to portal hypertension, such as esophageal varices rupture, is based on the use of ß-blockers, but some patients still present with acute variceal bleeding, mainly due to an insufficient reduction of portal pressure. In this study, we sought to demonstrate that the addition of folic acid to ß-blockers is more effective in reducing portal pressure than ß-blockers alone. This finding could represent the basis for validation studies in larger cohorts, which could impact the future prophylactic management of variceal bleeding in cirrhosis. Enhancing the benefit of ß-blockers with a safe, accessible, cost-effective drug could improve clinical outcomes in cirrhosis, which in turn could translate into a reduction in the rates and costs of hospitalization, and ultimately into improved survival.


Asunto(s)
Várices Esofágicas y Gástricas , Hipertensión Portal , Humanos , Propranolol/uso terapéutico , Propranolol/farmacología , Várices Esofágicas y Gástricas/complicaciones , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Presión Portal , Óxido Nítrico , Hemorragia Gastrointestinal/prevención & control , Antagonistas Adrenérgicos beta/uso terapéutico , Antagonistas Adrenérgicos beta/farmacología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/tratamiento farmacológico , Hipertensión Portal/etiología , Hipertensión Portal/complicaciones
2.
Alzheimers Res Ther ; 14(1): 134, 2022 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-36115980

RESUMEN

BACKGROUND AND OBJECTIVES: Vascular disease is a known risk factor for Alzheimer's disease (AD). Endothelial dysfunction has been linked to reduced cerebral blood flow. Endothelial nitric oxide synthase pathway (eNOS) upregulation is known to support endothelial health. This single-center, proof-of-concept study tested whether the use of three medications known to augment the eNOS pathway activity improves cognition and cerebral blood flow (CBF). METHODS: Subjects with mild AD or mild cognitive impairment (MCI) were sequentially treated with the HMG-CoA reductase synthesis inhibitor simvastatin (weeks 0-16), L-arginine (weeks 4-16), and tetrahydrobiopterin (weeks 8-16). The primary outcome of interest was the change in CBF as measured by MRI from baseline to week 16. Secondary outcomes included standard assessments of cognition. RESULTS: A total of 11 subjects were deemed eligible and enrolled. One subject withdrew from the study after enrollment, leaving 10 subjects for data analysis. There was a significant increase in CBF from baseline to week 8 by ~13% in the limbic and ~15% in the cerebral cortex. Secondary outcomes indicated a modest but significant increase in the MMSE from baseline (24.2±3.2) to week 16 (26.0±2.7). Exploratory analysis indicated that subjects with cognitive improvement (reduction of the ADAS-cog 13) had a significant increase in their respective limbic and cortical CBF. CONCLUSIONS: Treatment of mild AD/MCI subjects with medications shown to augment the eNOS pathway was well tolerated and associated with modestly increased cerebral blood flow and cognitive improvement. TRIAL REGISTRATION: This study is registered in https://www. CLINICALTRIALS: gov ; registration identifier: NCT01439555; date of registration submitted to registry: 09/23/2011; date of first subject enrollment: 11/2011.


Asunto(s)
Enfermedad de Alzheimer , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/tratamiento farmacológico , Arginina/farmacología , Arginina/uso terapéutico , Biopterinas/análogos & derivados , Circulación Cerebrovascular , Cognición , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Pruebas Neuropsicológicas , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Prueba de Estudio Conceptual , Simvastatina/farmacología , Simvastatina/uso terapéutico
3.
Nan Fang Yi Ke Da Xue Xue Bao ; 42(5): 718-723, 2022 May 20.
Artículo en Chino | MEDLINE | ID: mdl-35673916

RESUMEN

OBJECTIVE: To explore the therapeutic mechanism of tanshinone IIA in the treatment of pulmonary arterial hypertension (PAH) in rats. METHODS: A total of 100 male SD rats were randomized into 5 groups (n=20), and except for those in the control group with saline injection, all the rats were injected with monocrotaline (MCT) on the back of the neck to establish models of pulmonary hypertension. Two weeks after the injection, the rat models received intraperitoneal injections of tanshinone IIA (10 mg/kg), phosphatidylinositol 3 kinase (PI3K) inhibitor (1 mg/kg), both tanshinone IIA and PI3K inhibitor, or saline (model group) on a daily basis. After 2 weeks of treatment, HE staining and α-SMA immunofluorescence staining were used to evaluate the morphology of the pulmonary vessels of the rats. The phosphorylation levels of PI3K, protein kinase B (PKB/Akt) and endothelial nitric oxide synthase (eNOS) in the lung tissue were determined with Western blotting; the levels of eNOS and NO were measured using enzyme-linked immunosorbent assay (ELISA). RESULTS: The results of HE staining and α-SMA immunofluorescence staining showed that tanshinone IIA effectively inhibited MCT-induced pulmonary artery intimamedia thickening and muscularization of the pulmonary arterioles (P < 0.01). The results of Western blotting showed that treatment with tanshinone IIA significantly increased the phosphorylation levels of PI3K, Akt and eNOS proteins in the lung tissue of PAH rats; ELISA results showed that the levels of eNOS and NO were significantly decreased in the rat models after tanshinone IIA treatment (P < 0.01). CONCLUSION: Treatment with tanshinone IIA can improve MCT-induced pulmonary hypertension in rats through the PI3K/Akt-eNOS signaling pathway.


Asunto(s)
Hipertensión Pulmonar , Monocrotalina , Abietanos , Animales , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/tratamiento farmacológico , Masculino , Monocrotalina/toxicidad , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasa/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar , Ratas , Ratas Sprague-Dawley , Transducción de Señal
4.
Int J Immunopathol Pharmacol ; 36: 3946320221111117, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35731855

RESUMEN

OBJECTIVE: Formononetin is a bioactive isoflavone that has numerous medicinal benefits. We explored the feasibility and its mechanism of formononetin on treating acute deep vein thrombosis (DVT) in rats. MATERIALS AND METHODS: Inferior vena cava (IVC) stenosis was performed to establish the DVT rat model. First, different doses of formononetin were used to observe the feasibility of formononetin on treating DVT. In sham and DVT groups, rats were orally treated with vehicle. In the remaining groups, formononetin (10 mg/kg, 20 mg/kg, and 40 mg/kg) was orally treated once a day for 7 days at 24 h after IVC. After 7 days, the levels of thrombosis and inflammation related factors in plasma were measured. The expression of endothelial nitric oxide synthase (eNOS) was analyzed by western blot and immunofluorescence. Molecular docking was used to evaluate the interaction between the formononetin and eNOS. Further, the NOS inhibitor (L-NAME) was used to explore the mechanism of formononetin for DVT. RESULT: After treatment with formononetin, the average weights of thrombosis were decreased, and the levels of thrombosis and inflammation related factors were also significantly decreased. Additionally, phosphorylation of eNOS was increased with the formononetin administration. There is a good activity of formononetin to eNOS (total score = -6.8). However, the effects of 40 mg/kg formononetin were concealed by the NOS inhibitor (L-NAME). CONCLUSION: Formononetin reduces vascular endothelium injury induced by DVT through increasing eNOS in rats, which provides a potential drug for treatment of venous thrombosis.


Asunto(s)
Isoflavonas , Trombosis de la Vena , Animales , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Inflamación/metabolismo , Isoflavonas/metabolismo , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Simulación del Acoplamiento Molecular , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Ratas , Ratas Sprague-Dawley , Trombosis de la Vena/tratamiento farmacológico
5.
Small ; 18(17): e2200038, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35332670

RESUMEN

Tumor microenvironment (TME)-responsive intelligent photodynamic therapy (PDT) systems have attracted increasing interest in anticancer therapy, due to their potential to address significant and unsatisfactory therapeutic issues, such as limited tissue penetration, inevitable normal tissue damage, and excessive impaired vessels. Here, an H2 O2 -triggered intelligent LCL/ZnO PDT nanodelivery system is elaborately designed. LCL/ZnO can selectively regulate tumor-derived endothelial cells (TECs) and specifically kill tumor cells, by responding to different H2 O2 gradients in TECs and tumor cells. The LCL/ZnO is able to normalize tumor vessels, thereby resulting in decreased metastases, and ameliorating the immunosuppressive TME. Further analysis demonstrates that singlet oxygen (1 O2 )-activated transient receptor potential vanilloid-4-endothelial nitric oxide synthase signals generated in TECs by LCL/ZnO induce tumor vascular normalization, which is identified as a novel mechanism contributing to the increased ability of PDT to promote cancer therapy. In conclusion, designing an intelligent PDT nanodelivery system response to the TME, that includes both selective TECs regulation and specific tumor-killing, will facilitate the development of effective interventions for future clinical applications.


Asunto(s)
Neoplasias , Fotoquimioterapia , Óxido de Zinc , Línea Celular Tumoral , Células Endoteliales , Humanos , Neoplasias/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Fármacos Fotosensibilizantes/farmacología , Canales Catiónicos TRPV , Microambiente Tumoral
6.
Sheng Li Xue Bao ; 74(1): 93-109, 2022 Feb 25.
Artículo en Chino | MEDLINE | ID: mdl-35199130

RESUMEN

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.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , Nefropatías Diabéticas/tratamiento farmacológico , Endotelio Vascular , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Estrés Oxidativo , Ácido Peroxinitroso/metabolismo , Ácido Peroxinitroso/farmacología , Ácido Peroxinitroso/uso terapéutico
7.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-936368

RESUMEN

OBJECTIVE@#To explore the therapeutic mechanism of tanshinone IIA in the treatment of pulmonary arterial hypertension (PAH) in rats.@*METHODS@#A total of 100 male SD rats were randomized into 5 groups (n=20), and except for those in the control group with saline injection, all the rats were injected with monocrotaline (MCT) on the back of the neck to establish models of pulmonary hypertension. Two weeks after the injection, the rat models received intraperitoneal injections of tanshinone IIA (10 mg/kg), phosphatidylinositol 3 kinase (PI3K) inhibitor (1 mg/kg), both tanshinone IIA and PI3K inhibitor, or saline (model group) on a daily basis. After 2 weeks of treatment, HE staining and α-SMA immunofluorescence staining were used to evaluate the morphology of the pulmonary vessels of the rats. The phosphorylation levels of PI3K, protein kinase B (PKB/Akt) and endothelial nitric oxide synthase (eNOS) in the lung tissue were determined with Western blotting; the levels of eNOS and NO were measured using enzyme-linked immunosorbent assay (ELISA).@*RESULTS@#The results of HE staining and α-SMA immunofluorescence staining showed that tanshinone IIA effectively inhibited MCT-induced pulmonary artery intimamedia thickening and muscularization of the pulmonary arterioles (P < 0.01). The results of Western blotting showed that treatment with tanshinone IIA significantly increased the phosphorylation levels of PI3K, Akt and eNOS proteins in the lung tissue of PAH rats; ELISA results showed that the levels of eNOS and NO were significantly decreased in the rat models after tanshinone IIA treatment (P < 0.01).@*CONCLUSION@#Treatment with tanshinone IIA can improve MCT-induced pulmonary hypertension in rats through the PI3K/Akt-eNOS signaling pathway.


Asunto(s)
Animales , Masculino , Ratas , Abietanos , Hipertensión Pulmonar/tratamiento farmacológico , Monocrotalina/toxicidad , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Fosfatidilinositol 3-Quinasa/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar , Ratas Sprague-Dawley , Transducción de Señal
8.
Acta Physiologica Sinica ; (6): 93-109, 2022.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-927585

RESUMEN

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.


Asunto(s)
Humanos , Diabetes Mellitus , Nefropatías Diabéticas/tratamiento farmacológico , Endotelio Vascular , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Estrés Oxidativo , Ácido Peroxinitroso/uso terapéutico
9.
Acta Cir Bras ; 34(8): e201900802, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31618402

RESUMEN

PURPOSE: To reveal the function of miR-134 in myocardial ischemia. METHODS: Real-time PCR and western blotting were performed to measure the expression of miR-134, nitric oxide synthase 3 (NOS3) and apoptotic-associated proteins. Lactic dehydrogenase (LDH) assay, cell counting kit-8 (CCK-8), Hoechst 33342/PI double staining and flow cytometry assay were implemented in H9c2 cells, respectively. MiR-134 mimic/inhibitor was used to regulate miR-134 expression. Bioinformatic analysis and luciferase reporter assay were utilized to identify the interrelation between miR-134 and NOS3. Rescue experiments exhibited the role of NOS3. The involvement of PI3K/AKT was assessed by western blot analysis. RESULTS: MiR-134 was high regulated in the myocardial ischemia model, and miR-134 mimic/inhibitor transfection accelerated/impaired the speed of cell apoptosis and attenuated/exerted the cell proliferative prosperity induced by H/R regulating active status of PI3K/AKT signaling. LDH activity was also changed due to the different treatments. Moreover, miR-134 could target NOS3 directly and simultaneously attenuated the expression of NOS3. Co-transfection miR-134 inhibitor and pcDNA3.1-NOS3 highlighted the inhibitory effects of miR-134 on myocardial H/R injury. CONCLUSION: This present work puts insights into the crucial effects of the miR-134/NOS3 axis in myocardial H/R injury, delivering a potential therapeutic technology in future.


Asunto(s)
Hipoxia/metabolismo , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proliferación Celular/efectos de los fármacos , MicroARNs/genética , MicroARNs/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos
10.
Acta cir. bras ; 34(8): e201900802, 2019. tab, graf
Artículo en Inglés | LILACS | ID: biblio-1038128

RESUMEN

Abstract Purpose To reveal the function of miR-134 in myocardial ischemia. Methods Real-time PCR and western blotting were performed to measure the expression of miR-134, nitric oxide synthase 3 (NOS3) and apoptotic-associated proteins. Lactic dehydrogenase (LDH) assay, cell counting kit-8 (CCK-8), Hoechst 33342/PI double staining and flow cytometry assay were implemented in H9c2 cells, respectively. MiR-134 mimic/inhibitor was used to regulate miR-134 expression. Bioinformatic analysis and luciferase reporter assay were utilized to identify the interrelation between miR-134 and NOS3. Rescue experiments exhibited the role of NOS3. The involvement of PI3K/AKT was assessed by western blot analysis. Results MiR-134 was high regulated in the myocardial ischemia model, and miR-134 mimic/inhibitor transfection accelerated/impaired the speed of cell apoptosis and attenuated/exerted the cell proliferative prosperity induced by H/R regulating active status of PI3K/AKT signaling. LDH activity was also changed due to the different treatments. Moreover, miR-134 could target NOS3 directly and simultaneously attenuated the expression of NOS3. Co-transfection miR-134 inhibitor and pcDNA3.1-NOS3 highlighted the inhibitory effects of miR-134 on myocardial H/R injury. Conclusion This present work puts insights into the crucial effects of the miR-134/NOS3 axis in myocardial H/R injury, delivering a potential therapeutic technology in future.


Asunto(s)
Animales , Ratas , Daño por Reperfusión Miocárdica/metabolismo , MicroARNs/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Hipoxia/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , MicroARNs/genética , MicroARNs/uso terapéutico , Proliferación Celular/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Biomaterials ; 160: 69-81, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29396380

RESUMEN

Myocardial infarction (MI) leads to the mass death of cardiomyocytes accompanying with the unfavorable alternation of microenvironment, a fibrosis scar deprived of electrical communications, and the lack of blood supply in the infarcted myocardium. The three factors are inextricably intertwined and thus result in a conservative MI therapy efficacy in clinic. A holistic approach pertinently targeted to these three key points would be favorable to rebuild the heart functions. Here, an injectable conductive hydrogel was constructed via in situ Michael addition reaction between multi-armed conductive crosslinker tetraaniline-polyethylene glycol diacrylate (TA-PEG) and thiolated hyaluronic acid (HA-SH). The resultant soft conductive hydrogel with equivalent myocardial conductivity and anti-fatigue performance was loaded with plasmid DNA encoding eNOs (endothelial nitric oxide synthase) nanocomplexes and adipose derived stem cells (ADSCs) for treating MI. The TA-PEG/HA-SH/ADSCs/Gene hydrogel-based holistic system was injected into the infarcted myocardium of SD rats. We demonstrated an increased expression of eNOs in myocardial tissue the heightening of nitrite concentration, accompanied with upregulation of proangiogenic growth factors and myocardium related mRNA. The results of electrocardiography, cardiogram, and histological analysis convincingly revealed a distinct increase of ejection fraction (EF), shortened QRS interval, smaller infarction size, less fibrosis area, and higher vessel density, indicating a significant improvement of heart functions. This conception of combination approach by a conductive injectable hydrogel loaded with stem cells and gene-encoding eNOs nanoparticles will become a robust therapeutic strategy for the treatment of MI.


Asunto(s)
Portadores de Fármacos , Hidrogeles , Infarto del Miocardio/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III , Plásmidos , Animales , Terapia Genética/métodos , Humanos , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Plásmidos/genética , Plásmidos/uso terapéutico , Ratas , Ratas Sprague-Dawley
12.
Peptides ; 35(1): 78-85, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22465621

RESUMEN

Pulmonary vascular endothelial nitric oxide (NO) synthase (eNOS)-derived NO is the major stimulant of cyclic guanosine 5'-monophosphate (cGMP) production and NO/cGMP-dependent vasorelaxation in the pulmonary circulation. We recently synthesized multiple peptides and reported that an eleven amino acid (SSWRRKRKESS) peptide (P1) but not scrambled P1 stimulated the catalytic activity but not expression of eNOS and causes NO/cGMP-dependent sustained vasorelaxation in isolated pulmonary artery (PA) segments and in lung perfusion models. Since cGMP levels can also be elevated by inhibition of phosphodiesterase type 5 (PDE-5), this study was designed to test the hypothesis that P1-mediated vesorelaxation is due to its unique dual action as NO-releasing PDE-5 inhibitor in the pulmonary circulation. Treatment of porcine PA endothelial cells (PAEC) with P1 caused time-dependent increase in intracellular NO release and inhibition of the catalytic activity of cGMP-specific PDE-5 but not PDE-5 protein expression leading to increased levels of cGMP. Acute hypoxia-induced PA vasoconstriction ex vivo and continuous telemetry monitoring of hypoxia (10% oxygen)-induced elevated PA pressure in freely moving rats were significantly restored by administration of P1. Chronic hypoxia (10% oxygen for 4 weeks)-induced alterations in PA perfusion pressure, right ventricular hypertrophy, and vascular remodeling were attenuated by P1 treatment. These results demonstrate the potential therapeutic effects of P1 to prevent and/or arrest the progression of hypoxia-induced PAH via NO/cGMP-dependent modulation of hemodynamic and vascular remodeling in the pulmonary circulation.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Óxido Nítrico Sintasa de Tipo III/farmacología , Óxido Nítrico/metabolismo , Fragmentos de Péptidos/farmacología , Inhibidores de Fosfodiesterasa 5/farmacología , Vasodilatadores/farmacología , Secuencia de Aminoácidos , Animales , Presión Sanguínea/efectos de los fármacos , Hipoxia de la Célula , Células Cultivadas , GMP Cíclico/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Hipertrofia Ventricular Derecha/prevención & control , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Masculino , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Unión Proteica , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Ratas , Ratas Sprague-Dawley , Porcinos , Vasodilatadores/uso terapéutico
13.
Mol Biol Rep ; 37(7): 3067-72, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19908164

RESUMEN

Endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) may play an important role in attenuating cardiac remodeling and apoptosis after myocardial infarction. However, the anti-inflammation effects of eNOS in infarcted myocardium and the role of MAPK signaling in eNOS/NO mediated cardiac remodeling have not yet been elucidated. Adenovirus carrying Human eNOS gene was delivered locally into heart 4 days prior to induction of myocardial infarction (MI) by left anterior descending coronary artery ligation. Monocyte/macrophage infiltration was detected by ED-1 immunohistochemistry. Western blot was employed to examine the activation of MAPK. eNOS gene transfer significantly reduced myocardial infarct size and improved cardiac contractility as well as left ventricle (LV) diastolic function at 7 days after MI. In addition, eNOS gene transfer decreased monocyte/macrophage infiltration in the infarct region of the heart. Phosphorylation of MAPK after MI were also dramatically reduced by eNOS gene transfer. All the protective effects of eNOS were blocked by N(ω)-nitro-L-arginine methyl ester (L-NAME) administration, indicating a NO-mediated event. These results demonstrate that the eNOS/NO system provides cardiac protection after MI injury through inhibition of inflammation and suppression of MAPK signaling.


Asunto(s)
Técnicas de Transferencia de Gen , Sistema de Señalización de MAP Quinasas , Infarto del Miocardio/enzimología , Infarto del Miocardio/fisiopatología , Óxido Nítrico Sintasa de Tipo III/genética , Remodelación Ventricular/fisiología , Animales , Movimiento Celular , Ectodisplasinas/metabolismo , Terapia Genética , Humanos , Inflamación/complicaciones , Inflamación/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Masculino , Infarto del Miocardio/patología , Infarto del Miocardio/terapia , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Fosforilación , Ratas , Ratas Wistar , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Clin Invest Med ; 31(4): E206-11, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18682044

RESUMEN

PURPOSE: To analyze the association between Ki-67 and eNOS expression with the pathological grades of patients with intracranial ependymomas, and to determine its value in distinguishing the progression of the disease. METHODS: A clinicopathological study was undertaken in 82 patients with intracranial ependymomas. Tissue samples, obtained by tumour resection, were divided into three groups: low-grade, mid-grade and high-grade ependymomas. Tissue samples obtained from 15 patients with brain contusion were used as control. Immuno-histochemical staining was performed to analyze the association between Ki-67 and eNOS expression with various tumour grades. The cell proliferating marker Ki-67 was assessed by positive cell count. The levels of eNOS positive expression were evaluated as slight, moderate and intense. RESULTS: 48 of 82 cases (58.54%) expressed Ki-67 protein. Expression of Ki-67 and eNOS was negative in all control samples. Positive cell rates were 2.65+/-0.83 % in the low-grade, 9.63+/-0.08 % in the mid-grade, and 28.41+/-0.71 % in the high-grade ependymoma groups. In low-grade ependymomas there were 8 and 12 cases that expressed eNOS slightly or moderately. In the mid-grade ependymoma group eNOS was expressed moderately in 10 cases and intensely in 15. In the high-grade group 20 cases showed intense positive expression of eNOS. The Ki-67 positive cell counts for slight, moderate and intense eNOS expression were 2.20, 6.07 and 22.25, respectively. CONCLUSION: Ki-67 and eNOS expression in intracranial ependymoma tissue was associated with the histopathological grade and malignant degree.


Asunto(s)
Neoplasias Encefálicas , Ependimoma , Regulación de la Expresión Génica/efectos de los fármacos , Antígeno Ki-67/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/uso terapéutico , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Ependimoma/tratamiento farmacológico , Ependimoma/patología , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento
15.
Curr Pharm Des ; 11(31): 4059-67, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16378511

RESUMEN

Basic science research on erectile physiology has been devoted to investigating the pathogenesis of erectile dysfunction (ED) and has led to the conclusion that ED is predominately a disease of vascular origin. It is well recognized that the incidence of ED dramatically increases in men who suffer from diabetes mellitus, hypercholesterolemia, and cardiovascular disease. Endothelial nitric oxide synthase (eNOS) is an important factor in cardiovascular homeostasis, angiogenesis, and erectile function. Given the impact of endothelial-derived nitric oxide (NO) in vascular biology, a great deal of research over the past decade has focused on the role of NO synthesis from the endothelium in normal erectile physiology as well as in disease states. Loss of the functional integrity of the endothelium and subsequent endothelial dysfunction plays an integral role in the occurrence of ED. Therefore, a likely target of gene therapy for the treatment of ED is eNOS. This communication reviews the role of eNOS in erectile physiology and discusses the alterations in eNOS expression in various vascular diseases of the penis. Putative gene therapy interventions to restore eNOS expression and subsequent endothelial function may represent an exciting new therapeutic strategy for the future treatment of ED.


Asunto(s)
Disfunción Eréctil/terapia , Terapia Genética/métodos , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Disfunción Eréctil/etiología , Disfunción Eréctil/fisiopatología , Humanos , Masculino , Óxido Nítrico Sintasa de Tipo III/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA