Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Antioxidants (Basel) ; 10(3)2021 Mar 16.
Article in English | MEDLINE | ID: mdl-33809405

ABSTRACT

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.
Sci Rep ; 8(1): 13258, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30185869

ABSTRACT

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.


Subject(s)
Muscle, Smooth, Vascular/cytology , Neointima/metabolism , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Aminopropionitrile/pharmacology , Animals , Cell Proliferation , Cells, Cultured , Culture Media, Conditioned , Humans , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic , Up-Regulation
4.
FASEB J ; 31(9): 3787-3799, 2017 09.
Article in English | MEDLINE | ID: mdl-28522596

ABSTRACT

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.


Subject(s)
Angiotensin II/pharmacology , Cardiomegaly/chemically induced , Gene Expression Regulation, Enzymologic/physiology , Protein-Lysine 6-Oxidase/metabolism , Ventricular Remodeling/physiology , Animals , Cardiomegaly/enzymology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Inflammation , Mice , Mice, Transgenic , Myocardium/cytology , Protein-Lysine 6-Oxidase/genetics , Signal Transduction
5.
Clín. investig. arterioscler. (Ed. impr.) ; 28(6): 271-280, nov.-dic. 2016. ilus, graf
Article in Spanish | IBECS | ID: ibc-158497

ABSTRACT

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


Subject(s)
Humans , Inflammation/physiopathology , Aortic Aneurysm, Abdominal/physiopathology , Inflammation Mediators/analysis , Extracellular Matrix , SOX9 Transcription Factor/physiology , Immunohistochemistry/methods , Polymerase Chain Reaction , Lentivirus
6.
Clin Investig Arterioscler ; 28(6): 271-280, 2016.
Article in Spanish | MEDLINE | ID: mdl-27692634

ABSTRACT

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.


Subject(s)
Aortic Aneurysm, Abdominal/pathology , Extracellular Matrix Proteins/genetics , Inflammation/pathology , SOX9 Transcription Factor/genetics , Aorta/metabolism , Aortic Aneurysm, Abdominal/genetics , Blotting, Western , Case-Control Studies , Chromatin Immunoprecipitation/methods , Down-Regulation , Extracellular Matrix/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transfection , Tumor Necrosis Factor-alpha/metabolism
7.
Sci Rep ; 6: 25944, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27181368

ABSTRACT

Recent works have highlighted the role of NOR-1 in both smooth and skeletal muscle, and have proposed this nuclear receptor as a nexus that coordinates muscle performance and metabolic capacity. However, no muscle specific genes regulated by NOR-1 have been identified so far. To identify NOR-1 target genes, we over-expressed NOR-1 in human vascular smooth muscle cells (VSMC). These cells subjected to sustained over-expression of supraphysiological levels of NOR-1 experienced marked phenotypic changes and up-regulated the skeletal muscle protein X-linked (SMPX), a protein typically expressed in striated muscle and associated to cell shape. By transcriptional studies and DNA-protein binding assays, we identified a non-consensus NBRE site in human SMPX promoter, critical for NOR-1 responsiveness. The expression of SMPX was higher in human skeletal muscle myoblasts (HSMM) than in human VSMC, and further increased in HSMM differentiated to myotubes. NOR-1 silencing prevented SMPX expression in HSMM, as well as their differentiation to myotubes, but the up-regulation of SMPX was dispensable for HSMM differentiation. Our results indicate that NOR-1 regulate SMPX in human muscle cells and acts as a muscle regulatory factor, but further studies are required to unravel its role in muscle differentiation and hypertrophy.


Subject(s)
Membrane Transport Proteins/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Proteins/genetics , Muscle Proteins/metabolism , Binding Sites , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Humans , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/chemistry , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Promoter Regions, Genetic , Transcriptional Activation
8.
Dis Model Mech ; 9(5): 541-52, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26989193

ABSTRACT

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.


Subject(s)
Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/enzymology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/biosynthesis , Angiotensin II , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Biomarkers/metabolism , Collagen/metabolism , Disease Models, Animal , Disease Progression , Elastin/metabolism , Enzyme Induction/drug effects , Female , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Inflammation/pathology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Middle Aged , Severity of Illness Index , Up-Regulation/drug effects
9.
PLoS One ; 10(11): e0141932, 2015.
Article in English | MEDLINE | ID: mdl-26600038

ABSTRACT

Neuron-derived orphan receptor-1 (NOR-1) plays a major role in vascular biology by controlling fibroproliferative and inflammatory responses. Because microRNAs (miRNAs) have recently emerged as key players in the regulation of gene expression in the vasculature, here we have investigated the regulation of NOR-1 by miRNAs in endothelial cells. Computational algorithms suggest that NOR-1 could be targeted by members of the miR-17 family. Accordingly, ectopic over-expression of miR-17 or miR-20a in endothelial cells using synthetic premiRNAs attenuated the up-regulation of NOR-1 expression induced by VEGF (as evidenced by real time PCR, Western blot and immunocitochemistry). Conversely, the antagonism of these miRNAs by specific antagomirs prevented the down-regulation of NOR-1 promoted by miR-17 or miR-20a in VEGF-stimulated cells. Disruption of the miRNA-NOR-1 mRNA interaction using a custom designed target protector evidenced the selectivity of these responses. Further, luciferase reporter assays and seed-sequence mutagenesis confirmed that miR-17 and -20a bind to NOR-1 3'-UTR. Finally, miR-17 and -20a ameliorated the up-regulation of VCAM-1 mediated by NOR-1 in VEGF-stimulated cells. Therefore, miR-17 and -20a target NOR-1 thereby regulating NOR-1-dependent gene expression.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 3/genetics , 3' Untranslated Regions/genetics , Base Sequence , Binding Sites , Computational Biology , Down-Regulation/genetics , Humans , MicroRNAs/genetics , Molecular Sequence Data , Nuclear Receptor Subfamily 4, Group A, Member 3/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Thromb Haemost ; 113(6): 1323-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25809189

ABSTRACT

Matrix metalloproteinases (MMPs) are associated with tissue remodelling and repair. In non-vascular tissues, NR4A receptors have been involved in the regulation of MMPs by transcriptional repression mechanisms. Here, we analyse alternative mechanisms involving NR4A receptors in the modulation of MMP activity in vascular smooth muscle cells (VSMC). Lentiviral overexpression of NR4A receptors (NOR-1, Nurr1 and Nur77) in human VSMC strongly decreased MMP-2 and MMP-9 activities (analysed by zymography and DQ-gelatin assays) and protein levels. NR4A receptors also down-regulated MMP-2 mRNA levels. Real-time PCR analysis evidenced that alpha-2-macroglobulin (A2M), but not other MMP inhibitors (TIMP-1 and TIMP-2) were up-regulated in NR4A-transduced cells. Interestingly, A2M was expressed in human vascular tissues including the smooth muscle media layer. While NR4A receptors increased A2M expression and secretion in VSMC, NR4A knockdown significantly reduced basal A2M expression in these cells. The direct transcriptional regulation of the human A2M promoter by NR4A receptors was characterised in luciferase reporter assays, electrophoretic mobility shift assays and by chromatin immunoprecipitation, identifying a NGFI-B response element (NBRE-71/-64) essential for the NR4A-mediated induction. The blockade of A2M partially prevented the reduction of MMPs activity observed in NR4A-transduced cells. Although mouse A2M promoter was unresponsive to NR4A receptors, vascular MMP expression was attenuated in transgenic mice over-expressing human NOR-1 in VSMC challenged with lipopolysaccharide. Our results show that the pan-proteinase inhibitor A2M is expressed in the vasculature and that NR4A receptors modulate VSMC MMP activity by several mechanisms including the up-regulation of A2M.


Subject(s)
DNA-Binding Proteins/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , alpha-Macroglobulins/metabolism , Animals , Binding Sites , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression Regulation, Enzymologic , Humans , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Promoter Regions, Genetic , RNA Interference , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Transcription, Genetic , Transfection , alpha-Macroglobulins/genetics
11.
J Mol Cell Cardiol ; 80: 34-44, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25536180

ABSTRACT

Recent work has highlighted the role of NR4A receptors in atherosclerosis and inflammation. In vascular smooth muscle cell (VSMC) proliferation, however, NOR-1 (neuron-derived orphan receptor-1) exerts antagonistic effects to Nur77 and Nurr1. The aim of this study was to analyse the effect of NOR-1 in VSMC inflammatory response. We assessed the consequence of a gain-of-function of this receptor on the response of VSMC to inflammatory stimuli. In human VSMC, lentiviral over-expression of NOR-1 reduced lipopolysaccharide (LPS)-induced up-regulation of cytokines (IL-1ß, IL-6 and IL-8) and chemokines (MCP-1 and CCL20). Similar effects were obtained in cells stimulated with TNFα or oxLDL. Conversely, siRNA-mediated NOR-1 inhibition significantly increased the expression of pro-inflammatory mediators. Interestingly, in the aortas from transgenic mice that over-express human NOR-1 in VSMC (TgNOR-1), the up-regulation of cytokine/chemokine by LPS was lower compared to wild-type littermates. Similar results were obtained in VSMC from transgenic animals. NOR-1 reduced the transcriptional activity of NFκB sensitive promoters (in transient transfections), and the binding of NFκB to its responsive element (in electrophoretic mobility shift assays). Furthermore, NOR-1 prevented the activation of NFκB pathway by decreasing IκBα phosphorylation/degradation and inhibiting the phosphorylation and subsequent translocation of p65 to the nucleus (assessed by Western blot and immunocytochemistry). These effects were associated with an attenuated phosphorylation of ERK1/2, p38 MAPK and Jun N-terminal kinase, pathways involved in the activation of NFκB. In mouse challenged with LPS, the activation of the NFκB signalling was also attenuated in the aorta from TgNOR-1. Our data support a role for NOR-1 as a negative modulator of the acute response elicited by pro-inflammatory stimuli in the vasculature.


Subject(s)
DNA-Binding Proteins/metabolism , Inflammation/metabolism , Membrane Transport Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation Mediators/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Protein Binding , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Signal Transduction , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...