Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
2.
Antioxidants (Basel) ; 10(3)2021 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-33809405

RESUMO

Abdominal aortic aneurysm (AAA) is a common life-threatening condition characterized by exacerbated inflammation and the generation of reactive oxygen species. Pharmacological treatments to slow AAA progression or to prevent its rupture remain a challenge. Targeting phosphodiesterase 4 (PDE4) has been verified as an effective therapeutic strategy for an array of inflammatory conditions; however, no studies have assessed yet PDE4 in AAA. Here, we used angiotensin II (AngII)-infused apolipoprotein E deficient mice to study the involvement of the PDE4 subfamily in aneurysmal disease. PDE4B but not PDE4D was upregulated in inflammatory cells from both experimental and human AAA. The administration of the PDE4 selective inhibitor rolipram (3 mg/kg/day) to AngII-challenged mice (1000 ng/kg bodyweight/min) protected against AAA formation, limiting the progressive increase in the aortic diameter without affecting the blood pressure. The drug strongly attenuated the rise in vascular oxidative stress (superoxide anion) induced by AngII, and decreased the expression of inflammatory markers, as well as the recruitment of macrophages (MAC3+), lymphocytes (CD3+), and neutrophils (ELANE+) into the vessel wall. Rolipram also normalized the vascular MMP2 expression and MMP activity, preserving the elastin integrity and improving the vascular remodelling. These results point to PDE4B as a new therapeutic target for AAA.

3.
Int J Mol Sci ; 21(11)2020 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-32531895

RESUMO

Pulmonary arterial hypertension (PAH) is a life-threatening disease characterized by obstructed pulmonary vasculatures. Current therapies for PAH are limited and only alleviate symptoms. Reduced levels of BMPR2 are associated with PAH pathophysiology. Moreover, reactive oxygen species, inflammation and autophagy have been shown to be hallmarks in PAH. We previously demonstrated that MnTBAP, a synthetic metalloporphyrin with antioxidant and anti-inflammatory activity, inhibits the turn-over of BMPR2 in human umbilical vein endothelial cells. Therefore, we hypothesized that MnTBAP might be used to treat PAH. Human pulmonary artery endothelial cells (PAECs), as well as pulmonary microvascular endothelial (MVECs) and smooth muscle cells (MVSMCs) from PAH patients, were treated with MnTBAP. In vivo, either saline or MnTBAP was given to PAH rats induced by Sugen 5416 and hypoxia (SuHx). On PAECs, MnTBAP was found to increase BMPR2 protein levels by blocking autophagy. Moreover, MnTBAP increased BMPR2 levels in pulmonary MVECs and MVSMCs isolated from PAH patients. In SuHx rats, MnTBAP reduced right ventricular (RV) afterload by reversing pulmonary vascular remodeling, including both intima and media layers. Furthermore, MnTBAP improved RV function and reversed RV dilation in SuHx rats. Taken together, these data highlight the importance of MnTBAP as a potential therapeutic treatment for PAH.


Assuntos
Metaloporfirinas/farmacologia , Hipertensão Arterial Pulmonar/tratamento farmacológico , Hipertensão Arterial Pulmonar/fisiopatologia , Remodelação Vascular/efeitos dos fármacos , Animais , Autofagia/efeitos dos fármacos , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Testes de Função Cardíaca , Humanos , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Hipertensão Arterial Pulmonar/induzido quimicamente , Artéria Pulmonar/citologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/patologia , Ratos Sprague-Dawley
4.
Clín. investig. arterioscler. (Ed. impr.) ; 31(4): 166-177, jul.-ago. 2019. ilus
Artigo em Espanhol | IBECS | ID: ibc-182711

RESUMO

El aneurisma de aorta abdominal (AAA) es una patología vascular con una elevada tasa de morbimortalidad y una prevalencia que, en varones de más de 65 años, puede alcanzar el 8%. En esta enfermedad, habitualmente asintomática, se produce una dilatación progresiva de la pared vascular que puede llevar a su rotura, un fenómeno mortal en más de un 80% de los casos. El tratamiento de los pacientes con aneurismas asintomáticos se limita al seguimiento periódico con pruebas de imagen, el control de los factores de riesgo cardiovascular y un tratamiento con terapia antiagregante y estatinas, si bien actualmente no existe ningún tratamiento farmacológico efectivo capaz de limitar su progresión o evitar su rotura. En la actualidad el diámetro aórtico es el único marcador de riesgo de rotura y determina la necesidad de reparación quirúrgica cuando alcanza valores superiores a 5,5 cm. En esta revisión se tratan los principales aspectos relacionados con la epidemiología, los factores de riesgo, el diagnóstico y el manejo terapéutico del AAA, se exponen las dificultades para disponer de buenos biomarcadores de esta enfermedad y se describen las estrategias para la identificación de nuevas dianas terapéuticas y biomarcadores en el AAA


Abdominal aortic aneurysm (AAA) is a vascular pathology with a high rate of morbidity and mortality and a prevalence that, in men over 65 years, can reach around 8%. In this disease, usually asymptomatic, there is a progressive dilatation of the vascular wall that can lead to its rupture, a fatal phenomenon in more than 80% of cases. The treatment of patients with asymptomatic aneurysms is limited to periodic monitoring with imaging tests, control of cardiovascular risk factors and treatment with statins and antiplatelet therapy. There is no effective pharmacological treatment capable of limiting AAA progression or avoiding their rupture. At present, the aortic diameter is the only marker of risk of rupture and determines the need for surgical repair when it reaches values greater than 5.5 cm. This review addresses the main aspects related to epidemiology, risk factors, diagnosis and clinical management of AAA, exposes the difficulties to have good biomarkers of this pathology and describes the strategies for the identification of new therapeutic targets and biomarkers in AAA


Assuntos
Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Aneurisma da Aorta Abdominal/epidemiologia , Aneurisma da Aorta Abdominal/fisiopatologia , Biomarcadores/análise , Sistemas de Liberação de Medicamentos/métodos , Aneurisma da Aorta Abdominal/terapia , Hipertensão/complicações , Uso de Tabaco/epidemiologia , Doença Arterial Periférica/fisiopatologia , Inibidores da Enzima Conversora de Angiotensina/administração & dosagem
5.
Clin Investig Arterioscler ; 31(4): 166-177, 2019.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-30528271

RESUMO

Abdominal aortic aneurysm (AAA) is a vascular pathology with a high rate of morbidity and mortality and a prevalence that, in men over 65 years, can reach around 8%. In this disease, usually asymptomatic, there is a progressive dilatation of the vascular wall that can lead to its rupture, a fatal phenomenon in more than 80% of cases. The treatment of patients with asymptomatic aneurysms is limited to periodic monitoring with imaging tests, control of cardiovascular risk factors and treatment with statins and antiplatelet therapy. There is no effective pharmacological treatment capable of limiting AAA progression or avoiding their rupture. At present, the aortic diameter is the only marker of risk of rupture and determines the need for surgical repair when it reaches values greater than 5.5cm. This review addresses the main aspects related to epidemiology, risk factors, diagnosis and clinical management of AAA, exposes the difficulties to have good biomarkers of this pathology and describes the strategies for the identification of new therapeutic targets and biomarkers in AAA.


Assuntos
Aneurisma da Aorta Abdominal/fisiopatologia , Biomarcadores/metabolismo , Terapia de Alvo Molecular , Idoso , Aneurisma da Aorta Abdominal/diagnóstico , Aneurisma da Aorta Abdominal/terapia , Ruptura Aórtica/prevenção & controle , Progressão da Doença , Feminino , Humanos , Masculino , Prevalência , Fatores de Risco
6.
Sci Rep ; 8(1): 13258, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30185869

RESUMO

Lysyl oxidase (LOX) plays a critical role in extracellular matrix maturation and limits VSMC proliferation and vascular remodeling. We have investigated whether this anti-proliferative effect relies on the extracellular catalytically active LOX or on its biologically active propeptide (LOX-PP). High expression levels of both LOX and LOX-PP were detected in the vascular wall from transgenic mice over-expressing the full-length human LOX cDNA under the control of SM22α promoter (TgLOX), which targets the transgene to VSMC without affecting the expression of mouse LOX isoenzymes. TgLOX VSMC also secrete high amounts of both mature LOX and LOX-PP. Wild-type (WT) mouse VSMC exposed to VSMC supernatants from transgenic animals showed reduced proliferative rates (low [3H]-thymidine uptake and expression of PCNA) than those incubated with conditioned media from WT cells, effect that was abrogated by ß-aminopropionitrile (BAPN), an inhibitor of LOX activity. Lentiviral over-expression of LOX, but not LOX-PP, decreased human VSMC proliferation, effect that was also prevented by BAPN. LOX transgenesis neither impacted local nor systemic inflammatory response induced by carotid artery ligation. Interestingly, in this model, BAPN normalized the reduced neointimal thickening observed in TgLOX mice. Therefore, extracellular enzymatically active LOX is required to limit both VSMC proliferation and vascular remodeling.


Assuntos
Músculo Liso Vascular/citologia , Neointima/metabolismo , Proteína-Lisina 6-Oxidase/genética , Proteína-Lisina 6-Oxidase/metabolismo , Aminopropionitrilo/farmacologia , Animais , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados , Humanos , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Regiões Promotoras Genéticas , Regulação para Cima
7.
FASEB J ; 32(8): 4459-4469, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29547702

RESUMO

Vascular smooth muscle cells (VSMCs) transdifferentiate into osteoblast-like cells during vascular calcification, inducing active remodeling and calcification of the extracellular matrix (ECM). Intracellular and extracellular enzymes, such as lysyl hydroxylase 1 (PLOD1) and lysyl oxidase (LOX), contribute to ECM maturation and stabilization. We assessed the contribution of these enzymes to hyperphosphatemia-induced calcification. Human and murine VSMCs were differentiated into functional osteoblast-like cells by high-phosphate medium (HPM) conditioning. HPM promoted ECM calcification and up-regulated osteoblast markers associated with induction of LOX and PLOD1 expression and with an increase in ECM-insoluble collagen deposition. Murine VSMCs from transgenic mice overexpressing LOX (TgLOX) exhibited an increase in HPM-dependent calcification and osteoblast commitment compared with wild-type cells. Similarly, enhanced HPM-induced calcification was detected in aorta from TgLOX. Conversely, ß-aminopropionitrile (a LOX inhibitor) and LOX knockdown abrogated VSMC calcification and transdifferentiation. We found a significant positive association between LOX expression and vascular calcification in human atherosclerotic lesions. Likewise, 2,2'-dipyridil (a PLOD inhibitor) and PLOD1 knockdown impaired HPM-induced ECM mineralization and osteoblast commitment. Our findings identify LOX and PLOD as critical players in vascular calcification and highlight the importance of ECM remodeling in this process.-Jover, E., Silvente, A., Marín, F., Martínez-González, J., Orriols, M., Martinez, C. M., Puche, C. M., Valdés, M., Rodriguez, C., Hernández-Romero, D. Inhibition of enzymes involved in collagen cross-linking reduces vascular smooth muscle cell calcification.


Assuntos
Colágeno/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/fisiologia , Calcificação Vascular/metabolismo , Aminopropionitrilo/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/fisiologia , Transdiferenciação Celular/efeitos dos fármacos , Transdiferenciação Celular/fisiologia , Células Cultivadas , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/fisiologia , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Calcificação Vascular/tratamento farmacológico
8.
Front Physiol ; 8: 486, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28751863

RESUMO

Aim: Effective treatment of premature infants with bronchopulmonary dysplasia (BPD) is lacking. We hypothesize that bone morphogenetic protein 9 (BMP9), a ligand of the TGF-ß family that binds to the activin receptor-like kinase 1 (ALK1)-BMP receptor type 2 (BMPR2) receptor complex, may be a novel therapeutic option for BPD. Therefore, we investigated the cardiopulmonary effects of BMP9 in neonatal Wistar rats with hyperoxia-induced BPD. Methods: Directly after birth Wistar rat pups were exposed to 100% oxygen for 10 days. From day 2 rat pups received BMP9 (2.5 µg/kg, twice a day) or 0.9% NaCl by subcutaneous injection. Beneficial effects of BMP9 on aberrant alveolar development, lung inflammation and fibrosis, and right ventricular hypertrophy (RVH) were investigated by morphometric analysis and cytokine production. In addition, differential mRNA expression of BMP9 and its receptor complex: ALK1, BMPR2, and Endoglin, and of the ALK1 downstream target transmembrane protein 100 (TMEM100) were studied during the development of experimental BPD. Expression of the BMP9 receptor complex and TMEM100 was studied in human endothelial and epithelial cell cultures and the effect of BMP9 on inflammatory cytokine production and TMEM100 expression was studied in endothelial cell cultures. Results:ALK1, ALK2, BMPRII, TMEM100, and Endoglin were differentially expressed in experimental BPD, suggesting a role for BMP9-dependent signaling in the development of (experimental) BPD. TMEM100 was expressed in the wall of blood vessels, showing an elastin-like expression pattern in arterioles. Expression of TMEM100 mRNA and protein was decreased after exposure to hyperoxia. BMP9 treatment of rat pups with hyperoxia-induced experimental BPD reduced alveolar enlargement, lung septal thickness and fibrosis, and prevented inflammation, but did not attenuate vascular remodeling and RVH. The anti-inflammatory effect of BMP9 was confirmed in vitro. Highest expression of ALK1, BMPR2, and TMEM100 was observed in human endothelial cell cultures. Stimulation of human endothelial cell cultures with BMP9 reduced their pro-inflammatory cytokine response and induced TMEM100 expression in pulmonary arterial endothelial cells. Conclusion: BMP9 protects against neonatal hyperoxia-induced BPD by improving aberrant alveolar development, inflammation and fibrosis, demonstrating its therapeutic potential for premature infants with severe BPD.

9.
FASEB J ; 31(9): 3787-3799, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28522596

RESUMO

Lysyl oxidase (LOX) controls matrix remodeling, a key process that underlies cardiovascular diseases and heart failure; however, a lack of suitable animal models has limited our knowledge with regard to the contribution of LOX to cardiac dysfunction. Here, we assessed the impact of LOX overexpression on ventricular function and cardiac hypertrophy in a transgenic LOX (TgLOX) mouse model with a strong cardiac expression of human LOX. TgLOX mice exhibited high expression of the transgene in cardiomyocytes and cardiofibroblasts, which are associated with enhanced LOX activity and H2O2 production and with cardiofibroblast reprogramming. LOX overexpression promoted an age-associated concentric remodeling of the left ventricle and impaired diastolic function. Furthermore, LOX transgenesis aggravated angiotensin II (Ang II)-induced cardiac hypertrophy and dysfunction, which triggered a greater fibrotic response that was characterized by stronger collagen deposition and cross-linking and high expression of fibrotic markers. In addition, LOX transgenesis increased the Ang II-induced myocardial inflammatory infiltrate, exacerbated expression of proinflammatory markers, and decreased that of cardioprotective factors. Mechanistically, LOX overexpression enhanced oxidative stress and potentiated the Ang II-mediated cardiac activation of p38 MAPK while reducing AMPK activation. Our findings suggest that LOX induces an age-dependent disturbance of diastolic function and aggravates Ang II-induced hypertrophy, which provides novel insights into the role of LOX in cardiac performance.-Galán, M., Varona, S., Guadall, A., Orriols, M., Navas, M., Aguiló, S., de Diego, A., Navarro, M. A., García-Dorado, D., Rodríguez-Sinovas, A., Martínez-González, J., Rodriguez, C. Lysyl oxidase overexpression accelerates cardiac remodeling and aggravates angiotensin II-induced hypertrophy.


Assuntos
Angiotensina II/farmacologia , Cardiomegalia/induzido quimicamente , Regulação Enzimológica da Expressão Gênica/fisiologia , Proteína-Lisina 6-Oxidase/metabolismo , Remodelação Ventricular/fisiologia , Animais , Cardiomegalia/enzimologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Inflamação , Camundongos , Camundongos Transgênicos , Miocárdio/citologia , Proteína-Lisina 6-Oxidase/genética , Transdução de Sinais
11.
Cell Mol Life Sci ; 74(16): 2979-2995, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28447104

RESUMO

Pulmonary arterial hypertension (PAH) is a chronic disease characterized by a progressive elevation in mean pulmonary arterial pressure. This occurs due to abnormal remodeling of small peripheral lung vasculature resulting in progressive occlusion of the artery lumen that eventually causes right heart failure and death. The most common cause of PAH is inactivating mutations in the gene encoding a bone morphogenetic protein type II receptor (BMPRII). Current therapeutic options for PAH are limited and focused mainly on reversal of pulmonary vasoconstriction and proliferation of vascular cells. Although these treatments can relieve disease symptoms, PAH remains a progressive lethal disease. Emerging data suggest that restoration of BMPRII signaling in PAH is a promising alternative that could prevent and reverse pulmonary vascular remodeling. Here we will focus on recent advances in rescuing BMPRII expression, function or signaling to prevent and reverse pulmonary vascular remodeling in PAH and its feasibility for clinical translation. Furthermore, we summarize the role of described miRNAs that directly target the BMPR2 gene in blood vessels. We discuss the therapeutic potential and the limitations of promising new approaches to restore BMPRII signaling in PAH patients. Different mutations in BMPR2 and environmental/genetic factors make PAH a heterogeneous disease and it is thus likely that the best approach will be patient-tailored therapies.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/terapia , MicroRNAs/genética , Transdução de Sinais , Animais , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/análise , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , MicroRNAs/análise , MicroRNAs/metabolismo , Terapia de Alvo Molecular , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo
12.
Antioxid Redox Signal ; 27(7): 379-397, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28010122

RESUMO

AIMS: Vascular stiffness, structural elastin abnormalities, and increased oxidative stress are hallmarks of hypertension. Lysyl oxidase (LOX) is an elastin crosslinking enzyme that produces H2O2 as a by-product. We addressed the interplay between LOX, oxidative stress, vessel stiffness, and elastin. RESULTS: Angiotensin II (Ang II)-infused hypertensive mice and spontaneously hypertensive rats (SHR) showed increased vascular LOX expression and stiffness and an abnormal elastin structure. Mice over-expressing LOX in vascular smooth muscle cells (TgLOX) exhibited similar mechanical and elastin alterations to those of hypertensive models. LOX inhibition with ß-aminopropionitrile (BAPN) attenuated mechanical and elastin alterations in TgLOX mice, Ang II-infused mice, and SHR. Arteries from TgLOX mice, Ang II-infused mice, and/or SHR exhibited increased vascular H2O2 and O2.- levels, NADPH oxidase activity, and/or mitochondrial dysfunction. BAPN prevented the higher oxidative stress in hypertensive models. Treatment of TgLOX and Ang II-infused mice and SHR with the mitochondrial-targeted superoxide dismutase mimetic mito-TEMPO, the antioxidant apocynin, or the H2O2 scavenger polyethylene glycol-conjugated catalase (PEG-catalase) reduced oxidative stress, vascular stiffness, and elastin alterations. Vascular p38 mitogen-activated protein kinase (p38MAPK) activation was increased in Ang II-infused and TgLOX mice and this effect was prevented by BAPN, mito-TEMPO, or PEG-catalase. SB203580, the p38MAPK inhibitor, normalized vessel stiffness and elastin structure in TgLOX mice. INNOVATION: We identify LOX as a novel source of vascular reactive oxygen species and a new pathway involved in vascular stiffness and elastin remodeling in hypertension. CONCLUSION: LOX up-regulation is associated with enhanced oxidative stress that promotes p38MAPK activation, elastin structural alterations, and vascular stiffness. This pathway contributes to vascular abnormalities in hypertension. Antioxid. Redox Signal. 27, 379-397.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Hipertensão/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Transdução de Sinais , Rigidez Vascular , Animais , Modelos Animais de Doenças , Elastina/química , Proteínas da Matriz Extracelular/genética , Hipertensão/genética , Masculino , Camundongos , Estresse Oxidativo , Proteína-Lisina 6-Oxidase/genética , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Clín. investig. arterioscler. (Ed. impr.) ; 28(6): 271-280, nov.-dic. 2016. ilus, graf
Artigo em Espanhol | IBECS | ID: ibc-158497

RESUMO

Introducción: La fibulina-5 (FBLN5) es una proteína elastogénica implicada en el remodelado de la matriz extracelular (MEX), un proceso fundamental en el aneurisma de aorta abdominal (AAA). Sin embargo, no se ha determinado la posible contribución de la FBLN5 al AAA. Métodos: Se realizaron análisis por PCR a tiempo real, Western blot, transducción lentiviral, transfección transitoria e inmunoprecipitación de cromatina (ChIP) en aorta abdominal de pacientes con AAA o donantes y en células musculares lisas de aorta humana (CMLV). Resultados: La expresión vascular de la FBLN5 disminuye en la aorta abdominal de pacientes con AAA frente a donantes sanos. El nivel de ARNm y proteína de la FBLN5 y su secreción al espacio extracelular se redujeron en CMLV expuestas a estímulos inflamatorios. Este efecto se produce a través de un mecanismo transcripcional en el que está implicada una región proximal del promotor de la FBLN5 que contiene un elemento de respuesta a SOX. De hecho, la expresión de SOX9 se inhibe en CMLV tratadas con LPS y TNFα y disminuye en el AAA, en el que correlaciona con la de la FBLN5. Además, la sobreexpresión de SOX9 contrarrestó la disminución de la expresión y actividad transcripcional de la FBLN5 inducida por el TNFα. Finalmente, observamos que SOX9 interacciona con el promotor de la FBLN5 y que esta unión se reduce en respuesta a TNFα. Conclusiones: La inhibición de la FBLN5 en el AAA humano podría contribuir al remodelado destructivo de la matriz extracelular inducido por el componente inflamatorio de la patología


Introduction: Fibulin-5 (FBLN5) is an elastogenic protein critically involved in extracellular matrix (ECM) remodelling, a key process in abdominal aortic aneurysm (AAA). However, the possible contribution of FBLN5 to AAA development has not been addressed. Methods: Expression levels were determined by real-time PCR and Western blot in human abdominal aorta from patients with AAA or healthy donors, as well as in human aortic vascular smooth muscle cells (VSMC). Lentiviral transduction, transient transfections, and chromatin immunoprecipitation (ChIP) assays were also performed. Results: The expression of FBLN5 in human AAA was significantly lower than in healthy donors. FBLN5 mRNA and protein levels and their secretion to the extracellular environment were down-regulated in VSMC exposed to inflammatory stimuli. Interestingly, FBLN5 transcriptional activity was inhibited by TNFα and lipopolysaccharide (LPS), and depends on a SOX response element. In fact, SOX9 expression was reduced in VMSC induced by inflammatory mediators and in human AAA, and correlated with that of FBLN5. Furthermore, SOX9 over-expression limited the reduction of FBLN5 expression induced by cytokines in VSMC. Finally, it was observed that SOX9 interacts with FBLN5 promoter, and that this binding was reduced upon TNFα exposure. Conclusions: FBLN5 downregulation in human AAA could contribute to extracellular matrix remodelling induced by the inflammatory component of the disease


Assuntos
Humanos , Inflamação/fisiopatologia , Aneurisma da Aorta Abdominal/fisiopatologia , Mediadores da Inflamação/análise , Matriz Extracelular , Fatores de Transcrição SOX9/fisiologia , Imuno-Histoquímica/métodos , Reação em Cadeia da Polimerase , Lentivirus
14.
Clin Investig Arterioscler ; 28(6): 271-280, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-27692634

RESUMO

INTRODUCTION: Fibulin-5 (FBLN5) is an elastogenic protein critically involved in extracellular matrix (ECM) remodelling, a key process in abdominal aortic aneurysm (AAA). However, the possible contribution of FBLN5 to AAA development has not been addressed. METHODS: Expression levels were determined by real-time PCR and Western blot in human abdominal aorta from patients with AAA or healthy donors, as well as in human aortic vascular smooth muscle cells (VSMC). Lentiviral transduction, transient transfections, and chromatin immunoprecipitation (ChIP) assays were also performed. RESULTS: The expression of FBLN5 in human AAA was significantly lower than in healthy donors. FBLN5 mRNA and protein levels and their secretion to the extracellular environment were down-regulated in VSMC exposed to inflammatory stimuli. Interestingly, FBLN5 transcriptional activity was inhibited by TNFα and lipopolysaccharide (LPS), and depends on a SOX response element. In fact, SOX9 expression was reduced in VMSC induced by inflammatory mediators and in human AAA, and correlated with that of FBLN5. Furthermore, SOX9 over-expression limited the reduction of FBLN5 expression induced by cytokines in VSMC. Finally, it was observed that SOX9 interacts with FBLN5 promoter, and that this binding was reduced upon TNFα exposure. CONCLUSIONS: FBLN5 downregulation in human AAA could contribute to extracellular matrix remodelling induced by the inflammatory component of the disease.


Assuntos
Aneurisma da Aorta Abdominal/patologia , Proteínas da Matriz Extracelular/genética , Inflamação/patologia , Fatores de Transcrição SOX9/genética , Aorta/metabolismo , Aneurisma da Aorta Abdominal/genética , Western Blotting , Estudos de Casos e Controles , Imunoprecipitação da Cromatina/métodos , Regulação para Baixo , Matriz Extracelular/metabolismo , Humanos , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
15.
Cardiovasc Res ; 110(3): 431-42, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27089918

RESUMO

AIMS: Destructive remodelling of extracellular matrix (ECM) and inflammation lead to dilation and ultimately abdominal aortic aneurysm (AAA). Fibulin-5 (FBLN5) mediates cell-ECM interactions and elastic fibre assembly and is critical for ECM remodelling. We aimed to characterize FBLN5 regulation in human AAA and analyse the underlying mechanisms. METHODS AND RESULTS: FBLN5 expression was significantly decreased in human aneurysmatic aortas compared with healthy vessels. Local FBLN5 knockdown promoted aortic dilation and enhanced vascular expression of inflammatory markers in Ang II-infused C57BL/6J mice. Inflammatory stimuli down-regulated FBLN5 expression and transcriptional activity in human aortic vascular smooth muscle cells (VSMC). Further, aortic FBLN5 expression was reduced in LPS-challenged mice. A SOX response element was critical for FBLN5 promoter activity. The SOX9 expression pattern in human AAA parallels that of FBLN5, and like FBLN5, it was reduced in TNFα-stimulated VSMC. Interestingly, SOX9 over-expression prevented the cytokine-mediated reduction of FBLN5 expression and transcription. The inhibition of Class I histone deacetylases (HDACs) by MS-275 or gene silencing attenuated the inflammation-mediated decrease of FBLN5 expression in VSMC and in the vascular wall. Consistently, HDAC inhibition counteracted the reduction of SOX9 expression induced by inflammatory stimuli and prevented the TNFα-mediated decrease in the binding of SOX9 to FBLN5 promoter normalizing FBLN5 expression. CONCLUSION: We evidence the deregulation of FBLN5 in human AAA and identify a SOX9/HDAC-dependent mechanism involved in the down-regulation of FBLN5 by inflammation. HDAC inhibitors or pharmacological approaches that aimed to preserve FBLN5 could be useful to prevent the disorganization of ECM induced by inflammation in AAA.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Epigênese Genética , Proteínas da Matriz Extracelular/metabolismo , Mediadores da Inflamação/metabolismo , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Angiotensina II , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/induzido quimicamente , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/patologia , Sítios de Ligação , Estudos de Casos e Controles , Células Cultivadas , Dilatação Patológica , Modelos Animais de Doenças , Regulação para Baixo , Epigênese Genética/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Inibidores de Histona Desacetilases/farmacologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Regiões Promotoras Genéticas , Interferência de RNA , Proteínas Recombinantes/genética , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo , Transcrição Gênica , Transfecção , Fator de Necrose Tumoral alfa/metabolismo
16.
Dis Model Mech ; 9(5): 541-52, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26989193

RESUMO

Clinical management of abdominal aortic aneurysm (AAA) is currently limited to elective surgical repair because an effective pharmacotherapy is still awaited. Inhibition of histone deacetylase (HDAC) activity could be a promising therapeutic option in cardiovascular diseases. We aimed to characterise HDAC expression in human AAA and to evaluate the therapeutic potential of class I and IIa HDAC inhibitors in the AAA model of angiotensin II (Ang II)-infused apolipoprotein-E-deficient (ApoE(-/-)) mice. Real-time PCR, western blot and immunohistochemistry evidenced an increased expression of HDACs 1, 2 (both class I), 4 and 7 (both class IIa) in abdominal aorta samples from patients undergoing AAA open repair (n=22) compared with those from donors (n=14). Aortic aneurysms from Ang-II-infused ApoE(-/-) mice exhibited a similar HDAC expression profile. In these animals, treatment with a class I HDAC inhibitor (MS-275) or a class IIa inhibitor (MC-1568) improved survival, reduced the incidence and severity of AAA and limited aneurysmal expansion evaluated by Doppler ultrasonography. These beneficial effects were more potent in MC-1568-treated mice. The disorganisation of elastin and collagen fibres and lymphocyte and macrophage infiltration were effectively reduced by both inhibitors. Additionally, HDAC inhibition attenuated the exacerbated expression of pro-inflammatory markers and the increase in metalloproteinase-2 and -9 activity induced by Ang II in this model. Therefore, our data evidence that HDAC expression is deregulated in human AAA and that class-selective HDAC inhibitors limit aneurysm expansion in an AAA mouse model. New-generation HDAC inhibitors represent a promising therapeutic approach to overcome human aneurysm progression.


Assuntos
Aneurisma da Aorta Abdominal/tratamento farmacológico , Aneurisma da Aorta Abdominal/enzimologia , Inibidores de Histona Desacetilases/uso terapêutico , Histona Desacetilases/biossíntese , Angiotensina II , Animais , Aorta Abdominal/efeitos dos fármacos , Aorta Abdominal/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/metabolismo , Biomarcadores/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Elastina/metabolismo , Indução Enzimática/efeitos dos fármacos , Feminino , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Humanos , Inflamação/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Regulação para Cima/efeitos dos fármacos
17.
Thromb Haemost ; 112(4): 812-24, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24990180

RESUMO

Lysyl oxidase (LOX) is an extracellular matrix-modifying enzyme that seems to play a critical role in vascular remodelling. However, the lack of viable LOX-deficient animal models has been an obstacle to deep in LOX biology. In this study we have developed a transgenic mouse model that over-expresses LOX in vascular smooth muscle cells (VSMC) to clarify whether LOX could regulate VSMC phenotype and vascular remodelling. The SM22α proximal promoter drove the expression of a transgene containing the human LOX cDNA. Two stable transgenic lines, phenotypically indistinguishable, were generated by conventional methods (TgLOX). Transgene expression followed the expected SMC-specific pattern. In TgLOX mice, real-time PCR and immunohistochemistry evidenced a strong expression of LOX in the media from aorta and carotid arteries, coincident with a higher proportion of mature collagen. VSMC isolated from TgLOX mice expressed high levels of LOX pro-enzyme, which was properly secreted and processed into mature and bioactive LOX. Interestingly, cell proliferation was significantly reduced in cells from TgLOX mice. Transgenic VSMC also exhibited low levels of Myh10 (marker of SMC phenotypic switching), PCNA (marker of cell proliferation) and MCP-1, and a weak activation of Akt and ERK1/2 in response to mitogenic stimuli. Accordingly, neointimal thickening induced by carotid artery ligation was attenuated in TgLOX mice that also displayed a reduction in PCNA and MCP-1 immunostaining. Our results give evidence that LOX plays a critical role in vascular remodelling. We have developed a new animal model to study the role of LOX in vascular biology.


Assuntos
Proteína-Lisina 6-Oxidase/metabolismo , Remodelação Vascular/genética , Animais , Artérias Carótidas/patologia , Movimento Celular , Proliferação de Células , Quimiocina CCL2/metabolismo , Colágeno/química , Colágeno/metabolismo , DNA Complementar/metabolismo , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Cadeias Pesadas de Miosina/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Fenótipo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Regiões Promotoras Genéticas , Transfecção , Transgenes , Cicatrização
18.
Clín. investig. arterioscler. (Ed. impr.) ; 23(4): 168-174, jul.-ago. 2011. ilus
Artigo em Espanhol | IBECS | ID: ibc-92901

RESUMO

Introducción La lisil oxidasa (LOX), enzima implicada en la maduración de la matriz extracelular, juega un papel clave en el mantenimiento de la homeostasis del endotelio. Sin embargo, se desconoce el mecanismo a través del cual la LOX regula la función de la célula endotelial. Nuestro objetivo ha sido caracterizar los procesos celulares controlados por la LOX en células endoteliales. Métodos La LOX se sobreexpresó en células endoteliales humanas de vena de cordón umbilical (HUVEC) utilizando un sistema lentiviral. Las consecuencias de la sobreexpresión de LOX sobre el patrón de expresión se determinaron mediante microarrays. La expresión de LOX y de la (..) (AU)


Abstract Introduction: Lysyl oxidase (LOX) is an enzyme involved in extracellular matrix maturation that plays a crucial role in the maintenance of endothelial homeostasis. However, the specific mechanisms underlying the ability of LOX to regulate endothelial function are currently unknown. Our objective was to characterize the cellular processes controlled by LOX in endothelial cells. Methods: LOX was over expressed in human umbilical vein endothelial cells (HUVEC) when a lentiviral system was used. The consequences of LOX over expression were analyzed by microarray.LOX and 2-macroglobulin (A2 M) expression was assessed by real-time polymerase chain reaction and/or Western-Blot. Results: The lentiviral expression system significantly increased LOX expression in HUVEC (more than 20-fold). In agreement with this result, Western-Blot analysis revealed a marked enhancement of LOX protein levels in the cell extract for both the mature and catalytically active form and for the pro-enzyme. This approach also increased the level of LOX secreted into the culture medium. By microarray analysis we demonstrated that LOX over expression strongly alters the endothelial expression pattern and regulates genes mainly involved in the control of cell signaling, communication and adhesion. A2 M, a pan protease inhibitor with multiple biological activities, was one of the genes most strongly inhibited in LOX overexpressing cells. Conclusion: LOX overexpression strongly affects the expression profile of endothelial cells and significantly reduces A2M expression, an effect that could have a substantial impact on endothelial cell function (AU)


Assuntos
Humanos , Regulação da Expressão Gênica , Proteína-Lisina 6-Oxidase/farmacocinética , alfa-Macroglobulinas/análise , Células Endoteliais/fisiologia , Endotélio Vascular/fisiopatologia
19.
Front Biosci (Elite Ed) ; 3(3): 955-67, 2011 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622104

RESUMO

The adaptive response of endothelial cells to hypoxia involves a substantial remodeling of extracellular matrix (ECM). In endothelial cells hypoxia up-regulates lysyl oxidase (LOX), a key enzyme in ECM assembly, relevant to vascular homeostasis. However, the mechanism underlying this response has not been established. Hypoxia up-regulated LOX expression in endothelial cells (HUVEC and BAEC) and concomitantly increased LOX enzymatic activity. This effect was independent of autocrine factors released by hypoxic cells and relies on a transcriptional mechanism. Both mTOR blockade and HIF-1alpha knockdown slightly prevented LOX up-regulation by hypoxia, suggesting that HIF-1alpha is only partially responsible for this effect. In fact, serial promoter deletion and mutagenesis studies indicated a limited contribution of the previously described hypoxia response element (-75 bp). Interestingly, Smad over-expression further increased LOX transcriptional activity in endothelial cells exposed to hypoxia. Moreover, the increase in LOX expression triggered by hypoxia was significantly reduced by reactive oxygen species (ROS) inhibitors. Thus, our data support a role of Smad signaling and ROS in the up-regulation of LOX by hypoxia in endothelial cells.


Assuntos
Hipóxia Celular , Endotélio Vascular/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Regulação para Cima , Animais , Sequência de Bases , Western Blotting , Bovinos , Células Cultivadas , Primers do DNA , Endotélio Vascular/citologia , Indução Enzimática , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteína-Lisina 6-Oxidase/biossíntese , Proteína-Lisina 6-Oxidase/genética , Sirolimo/farmacologia , Transcrição Gênica
20.
J Lipid Res ; 52(4): 720-31, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21296955

RESUMO

Hypoxia affects vascular function and cell metabolism, survival, growth, and motility; these processes are partially regulated by prostanoids. We analyzed the effect of hypoxia and inflammation on key enzymes involved in prostanoid biosynthesis in human vascular cells. In human vascular smooth muscle cells (VSMC), hypoxia and interleukin (IL)-1ß synergistically increased prostaglandin (PG)I2 but not PGE2 release, thereby increasing the PGI2/PGE2 ratio. Concomitantly, these stimuli upregulated cyclooxygenase-2 (COX-2) expression (mRNA and protein) and COX activity. Interestingly, hypoxia enhanced PGI-synthase (PGIS) expression and activity in VSMC and human endothelial cells. Hypoxia did not significantly modify the inducible microsomal-PGE-synthase (mPGES)-1. Hypoxia-inducible factor (HIF)-1α-silencing abrogated hypoxia-induced PGIS upregulation. PGIS transcriptional activity was enhanced by hypoxia; however, the minimal PGIS promoter responsive to hypoxia (-131 bp) did not contain any putative hypoxia response element (HRE), suggesting that HIF-1 does not directly drive PGIS transcription. Serial deletion and site-directed mutagenesis studies suggested several transcription factors participate cooperatively. Plasma levels of the stable metabolite of PGI2 and PGIS expression in several tissues were also upregulated in mice exposed to hypoxia. These data suggest that PGIS upregulation is part of the adaptive response of vascular cells to hypoxic stress and could play a role in counteracting the deleterious effect of inflammatory stimuli.


Assuntos
Hipóxia Celular/fisiologia , Epoprostenol/metabolismo , Interleucina-1beta/farmacologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Western Blotting , Hipóxia Celular/genética , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/enzimologia , Células Endoteliais/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/efeitos dos fármacos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Prostaglandina-Endoperóxido Sintases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...