Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 53
Filtrer
1.
Braz J Cardiovasc Surg ; 38(6): e20220260, 2023 10 06.
Article de Anglais | MEDLINE | ID: mdl-37801489

RÉSUMÉ

INTRODUCTION: Thoracic aortic aneurysm is a potentially fatal disease with a strong genetic contribution. The dysfunction of vascular smooth muscle cells (VSMCs) contributes to the formation of this aneurysm. Although previous studies suggested that long non-coding ribonucleic acid (RNA) hypoxia inducible factor 1 α-antisense RNA 1 (HIF1A-AS1) exerted a vital role in the progression and pathogenesis of thoracic aortic aneurysm, we managed to find a new regulatory mechanism of HIF1A-AS1 in VSMCs via transcriptomics. METHODS: Cell viability was detected by the cell counting kit-8 assay. Cell apoptosis was assessed by Annexin V-fluorescein isothiocyanate/propidium iodide double staining. Transwell migration assay and wound healing assay were performed to check the migration ability of HIF1A-AS1 on VSMCs. The NextSeq XTen system (Illumina) was used to collect RNA sequencing data. Lastly, reverse transcription-quantitative polymerase chain reaction confirmed the veracity and reliability of RNA-sequencing results. RESULTS: We observed that overexpressing HIF1A-AS1 successfully promoted apoptosis, significantly altered cell cycle distribution, and greatly attenuated migration in VSMCs, further highlighting the robust promoting effects of HIF1A-AS1 to thoracic aortic aneurysm. Moreover, transcriptomics was implemented to uncover its underlying mechanism. A total of 175 differently expressed genes were identified, with some of them enriched in apoptosis, migration, and cell cycle-related pathways. Intriguingly, some differently expressed genes were noted in vascular development or coagulation function pathways. CONCLUSION: We suggest that HIF1A-AS1 mediated the progression of thoracic aortic aneurysm by not only regulating the function of VSMCs, but also altering vascular development or coagulation function.


Sujet(s)
Anévrysme de l'aorte thoracique , ARN long non codant , Humains , Anévrysme de l'aorte thoracique/génétique , microARN/génétique , Muscles lisses vasculaires , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Reproductibilité des résultats , Transcriptome , ARN long non codant/métabolisme
2.
Vet Pathol ; 59(5): 814-823, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35587717

RÉSUMÉ

Vascular mineralization is a hallmark of enzootic calcinosis. Histopathological, ultrastructural, and immunohistochemical investigations were performed on the external carotid arteries of seven sheep naturally poisoned by Nierembergia veitchii. Histologically, moderate to marked hyperplasia of the tunica intima was observed without mineralization. The tunica media exhibited mild to severe mineralization and osteochondroid metaplasia. Sheep with enzootic calcinosis showed arterial overexpression of osteopontin and tissue-nonspecific alkaline phosphatase and immunolabeling for osteonectin and osteocalcin in both intima and media layers of the tested arteries. The main ultrastructural finding in the tunica media was a marked phenotypic change of vascular smooth muscle cells from a contractile phenotype (VSMC-C) into a synthetic phenotype (VSMC-S). In the tunica media, VSMC-S produced matrix and extracellular vesicles, forming mineralizable granules associated with arterial mineralization. VSMC-S were also present in the tunica intima, but matrix and extracellular vesicles and mineralization were not observed. The absence of matrix and extracellular vesicles in the intimal hyperplasia, even in the presence of noncollagenous bone proteins, tissue-nonspecific alkaline phosphatase, and vitamin D receptors, reinforces the hypothesis that the presence of matrix and extracellular vesicles are crucial for the development of vascular mineralization in enzootic calcinosis. It is proposed that the two different VSMC-S phenotypes in calcinosis are due to the expression of at least two genetically different types of these cells induced by the action of 1,25(OH)2D3.


Sujet(s)
Calcinose , Hyperplasie , Maladies des ovins , Phosphatase alcaline/métabolisme , Animaux , Calcinose/médecine vétérinaire , Cellules cultivées , Hyperplasie/anatomopathologie , Hyperplasie/médecine vétérinaire , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Ovis , Maladies des ovins/anatomopathologie
3.
J Cardiovasc Pharmacol ; 77(1): 43-48, 2021 01 01.
Article de Anglais | MEDLINE | ID: mdl-33079831

RÉSUMÉ

ABSTRACT: Disruption of protein quality control occurs with aging and cardiovascular pathologies including arterial stiffness and hypertension. Angiotensin II (Ang II) is believed to induce endoplasmic reticulum stress in vascular smooth muscle cells (VSMCs), thus contributing to vascular remodeling and dysfunction. However, whether Ang II increases formation of protein aggregates and mediates proteotoxicity in VSMCs remain obscure. Accordingly, this study aimed to establish a quantitative method of protein aggregate detection induced by Ang II and to investigate their potential involvement in inflammatory and senescence responses. Proteostat staining showed increased aggregate numbers per cell on Ang II exposure. Immunoblot analysis further showed an increase in preamyloid oligomer presence in a detergent insoluble protein fraction purified from VSMCs stimulated with Ang II. Moreover, these responses were attenuated by treatment with chemical chaperone, 4-phenylbutyrate. 4-phenylbutyrate further blocked Ang II-induced senescence associated ß-galactosidase activity and THP-1 monocyte adhesion in VSMCs. These data suggest that Ang II induces proteotoxicity in VSMCs which likely contributes to aging and inflammation in the vasculature.


Sujet(s)
Angiotensine-II/toxicité , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Agrégats de protéines , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Humains , Mâle , Monocytes/effets des médicaments et des substances chimiques , Monocytes/métabolisme , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Agrégation pathologique de protéines , Rat Sprague-Dawley , Cellules THP-1
4.
Cell Biol Int ; 45(4): 882-889, 2021 Apr.
Article de Anglais | MEDLINE | ID: mdl-33377550

RÉSUMÉ

Prostate cancer is a life-threatening condition worldwide. As the tumor progresses, smooth muscle cells (SMCs) become atrophic/dedifferentiated, within a series of stromal changes named stromal reaction. Here, we tested whether a laminin 111-rich extracellular matrix (Lr-ECM) could affect SMCs phenotype and differentiation status. Using time-lapse microscopy, image analyses, quantitative real-time reverse transcription polymerase chain reaction, immunohistochemistry and immunoblotting, and transmission electron microscopy, we showed that SMCs acquires a migratory behavior with a decreased expression of differentiation markers and relocation of focal adhesion kinase. SMCs set homotypic cell junctions and were active in autophagy/phagocytosis. Analysis of the migratory behavior showed that SMCs polarized and migrated toward each other, recognizing long-distance signals such as matrix tensioning. However, half of the cell population were immotile, irrespective of the nearest neighbor distance, suggesting they do not engage in productive interactions, possibly as a result of back-to-back positioning. In conclusion, the Lr-ECM, mimics the effects of the proliferating and infiltrating tumor epithelium, causing SMCs phenotypical change similar to that observed in the stromal reaction, in addition to a hitherto undescribed, stereotyped pattern of cell motility resulting from cell polarization.


Sujet(s)
Myocytes du muscle lisse , Prostate , Tumeurs de la prostate , Animaux , Différenciation cellulaire , Mouvement cellulaire , Cellules cultivées , Matrice extracellulaire , Laminine/métabolisme , Mâle , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Prostate/métabolisme , Prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Rats , Rat Wistar
5.
Mol Hum Reprod ; 27(2)2021 02 05.
Article de Anglais | MEDLINE | ID: mdl-33313783

RÉSUMÉ

Experimental autoimmune orchitis (EAO) is a useful model to study organ-specific autoimmunity and chronic testicular inflammation. This model reflects testicular pathological changes reported in immunological infertility in men. Progression of EAO in rodents is associated with a significantly increased percentage of testicular endothelial cells and interstitial testicular blood vessels, indicating an ongoing angiogenic process. Vascular endothelial growth factor A (VEGFA), the main regulator of physiological and pathological angiogenesis, can stimulate endothelial cell proliferation, chemotaxis and vascular permeability. The aim of this study was to explore the role of VEGFA in the pathogenesis of testicular inflammation. Our results found VEGFA expression in Leydig cells, endothelial cells and macrophages in testis of rats with autoimmune orchitis. VEGFA level was significantly higher in testicular fluid and serum of rats at the end of the immunization period, preceding testicular damage. VEGF receptor (VEGFR) 1 is expressed mainly in testicular endothelial cells, whereas VEGFR2 was detected in germ cells and vascular smooth muscle cells. Both receptors were expressed in testicular interstitial cells. VEGFR2 increased after the immunization period in the testicular interstitium and VEGFR1 was downregulated in EAO testis. In-vivo-specific VEGFA inhibition by Bevacizumab prevented the increase in blood vessel number and reduced EAO incidence and severity. Our results unveil relevance of VEGFA-VEGFR axis during orchitis development, suggesting that VEGFA might be an early marker of testicular inflammation and Bevacizumab a therapeutic tool for treatment of testicular inflammation associated with subfertility and infertility.


Sujet(s)
Maladies auto-immunes/anatomopathologie , Néovascularisation pathologique , Testicule/vascularisation , Testicule/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Inhibiteurs de l'angiogenèse/pharmacologie , Animaux , Maladies auto-immunes/immunologie , Maladies auto-immunes/métabolisme , Maladies auto-immunes/prévention et contrôle , Bévacizumab/pharmacologie , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Cellules de Leydig/métabolisme , Cellules de Leydig/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Mâle , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Orchite/immunologie , Orchite/métabolisme , Orchite/prévention et contrôle , Caille/embryologie , Rat Wistar , Transduction du signal , Testicule/effets des médicaments et des substances chimiques , Récepteur-1 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme
6.
Eur J Pharmacol ; 887: 173438, 2020 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-32795515

RÉSUMÉ

Despite advances in medical therapy, pulmonary arterial hypertension (PAH) remains an inexorably progressive and highly lethal disease. Signal transducer and activator of transcription (STAT)-3 is one of the main intracellular transcription factors implicated in PAH vascular remodeling. We hypothesized that niclosamide, a STAT3 inhibitor, would reduce vascular remodeling in an established pulmonary arterial hypertension model, thus enhancing cardiac function. Male Wistar rats were treated either with monocrotaline (60 mg/kg), to induce PAH, or saline (C group) by intraperitoneal injection. On day 14, PAH animals were randomly assigned to receive oral (1) saline (PAH-SAL); (2) niclosamide (75 mg/kg/day) (PAH-NICLO); (3) sildenafil (20 mg/kg/day) (PAH-SIL); or (4) niclosamide + sildenafil (PAH-NICLO + SIL), once daily for 14 days. On day 28, right ventricular systolic pressure was lower in all treated groups compared to PAH-SAL. Pulmonary vascular collagen content was lower in PAH-NICLO (37 ± 3%) and PAH-NICLO + SIL (37 ± 6%) compared to PAH-SAL (68 ± 4%), but not in PAH-SIL (52 ± 1%). CD-34, an endothelial cell marker, was higher, while vimentin, a mesenchymal cell marker, was lower in PAH-NICLO and PAH-NICLO + SIL compared to PAH-SAL, suggesting attenuation of endothelial-mesenchymal transition. Expression of STAT3 downstream targets such as transforming growth factor (TGF)-ß, hypoxia-inducible factor (HIF)-1, and provirus integration site for Moloney murine leukemia virus (PIM-1) in lung tissue was reduced in PAH-NICLO and PAH-NICLO + SIL compared to PAH-SAL. In conclusion, niclosamide, with or without sildenafil, mitigated vascular remodeling and improved right ventricle systolic pressure. This new role for a well-established drug may represent a promising therapy for PAH.


Sujet(s)
Poumon/vascularisation , Poumon/effets des médicaments et des substances chimiques , Niclosamide/usage thérapeutique , Hypertension artérielle pulmonaire/prévention et contrôle , Remodelage vasculaire/effets des médicaments et des substances chimiques , Animaux , Cellules cultivées , Relation dose-effet des médicaments , Poumon/anatomopathologie , Mâle , Monocrotaline/toxicité , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Niclosamide/pharmacologie , Hypertension artérielle pulmonaire/induit chimiquement , Hypertension artérielle pulmonaire/anatomopathologie , Rats , Rat Wistar , Remodelage vasculaire/physiologie
7.
Biochim Biophys Acta Mol Basis Dis ; 1866(1): 165587, 2020 01 01.
Article de Anglais | MEDLINE | ID: mdl-31678158

RÉSUMÉ

Mechanisms whereby fibrillin-1 mutations determine thoracic aorta aneurysms/dissections (TAAD) in Marfan Syndrome (MFS) are unclear. Most aortic aneurysms evolve from mechanosignaling deregulation, converging to impaired vascular smooth muscle cell (VSMC) force-generating capacity accompanied by synthetic phenotype switch. However, little is known on VSMC mechanoresponses in MFS pathophysiology. Here, we investigated traction force-generating capacity in aortic VSMC cultured from 3-month old mg∆lpn MFS mice, together with morpho-functional and proteomic data. Cultured MFS-VSMC depicted marked phenotype changes vs. wild-type (WT) VSMC, with overexpressed cell proliferation markers but either lower (calponin-1) or higher (SM alpha-actin and SM22) differentiation marker expression. In parallel, the increased cell area and its complex non-fusiform shape suggested possible transition towards a mesenchymal-like phenotype, confirmed through several markers (e.g. N-cadherin, Slug). MFS-VSMC proteomic profile diverged from that of WT-VSMC particularly regarding lower expression of actin cytoskeleton-regulatory proteins. Accordingly, MFS-VSMC displayed lower traction force-generating capacity and impaired contractile moment at physiological substrate stiffness, and markedly attenuated traction force responses to enhanced substrate rigidity. Such impaired mechanoresponses correlated with decreased number, altered morphology and delocalization of focal adhesions, as well as disorganized actin stress fiber network vs. WT-VSMC. In VSMC cultured from 6-month-old mice, phenotype changes were attenuated and both WT-VSMC and MFS-VSMC generated less traction force, presumably involving VSMC aging, but without evident senescence. In summary, MFS-VSMC display impaired force-generating capacity accompanying a mesenchymal-like phenotype switch connected to impaired cytoskeleton/focal adhesion organization. Thus, MFS-associated TAAD involves mechanoresponse impairment common to other TAAD types, but through distinct mechanisms.


Sujet(s)
Syndrome de Marfan/anatomopathologie , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Actines/métabolisme , Animaux , Aorte/métabolisme , Aorte/anatomopathologie , Anévrysme de l'aorte/métabolisme , Anévrysme de l'aorte/anatomopathologie , Marqueurs biologiques/métabolisme , Différenciation cellulaire/physiologie , Prolifération cellulaire/physiologie , Cellules cultivées , Cytosquelette/métabolisme , Cytosquelette/anatomopathologie , Modèles animaux de maladie humaine , Femelle , Fibrilline-1/métabolisme , Contacts focaux/métabolisme , Contacts focaux/anatomopathologie , Mâle , Syndrome de Marfan/métabolisme , Souris , Souris de lignée C57BL , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Phénotype , Protéomique/méthodes
8.
Cell Signal ; 66: 109458, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31678252

RÉSUMÉ

The understanding of vascular calcification-based mechanism is an urgent pending task in vascular biology and this prompted us to better address this issue by investigating whether DNA methylation mechanism might drive osteogenic marker genes modulation in primary human vascular smooth muscle cells (VSMCs) responding to calcium and phosphate levels overload up to 72 h. Firstly, our data shows this calcifying process recapitulates the molecular repertory of osteogenic biomarkers and specifically requiring RUNX2, Osterix and ALP, BSP genes activations along 72 h in vitro, and this behavior was validated here using other lineages. Conversely, both BMPs 4 and 7 were significantly overexpressed, maybe already as a mechanism in response to RUNX2 and Osterix genes activities identified earlier in response to the calcifying condition, and taken into maintain the calcifying phenotype of VSMCs. Additionally, survival signaling was maintained active and accompanied by a dynamic cytoskeleton rearrangement signaling requiring MAPK and AKT phosphorylations. Moreover, during the contractile-to-calcifying transition phenotype of VSMCs, epigenetic machinery was finely modulated, requiring the translocation of DNMT3B and TET2 into nucleus and this prompted us evaluating whether the profile of osteogenic-related gene promoters' methylation might contribute with this process. By firstly estimating 5meC/5 hmeC ratio changes, we further specifically show the significance of the epigenetic modulation of Osterix and Bone sialoprotein related gene promoters, presenting a positive correlation between the epigenetic signature of their gene promoters and transcriptional patterns. Altogether, our results show for the first time the importance of epigenetic mechanism on modulating osteogenic gene markers reprogramming during calcifying VSMCs phenotype acquisition, which might drive the genesis of vascular ectopic calcification.


Sujet(s)
Épigenèse génétique , Myocytes du muscle lisse/anatomopathologie , Ostéogenèse , Calcification vasculaire/métabolisme , Phosphatase alcaline/métabolisme , Protéines morphogénétiques osseuses , Lignée cellulaire , Sous-unité alpha 1 du facteur CBF/métabolisme , Méthylation de l'ADN , Muscles lisses vasculaires/cytologie , Facteur de transcription Sp7/métabolisme
9.
Biol Res ; 52(1): 59, 2019 Dec 04.
Article de Anglais | MEDLINE | ID: mdl-31801629

RÉSUMÉ

OBJECTIVES: In varicose veins, vascular smooth muscle cells (VSMCs) often shows phenotypic transition and abnormal proliferation and migration. Evidence suggests the FOXC2-Notch pathway may be involved in the pathogenesis of varicose veins. Here, this study aimed to explore the role of long non-coding RNA FOXC2-AS1 (FOXC2 antisense RNA 1) in phenotypic transition, proliferation, and migration of varicose vein-derived VSMCs and to explore whether the FOXC2-Notch pathway was involved in this process. METHODS: The effect of FOXC2-AS1 on the proliferation and migration of human great saphenous vein smooth muscle cells (SV-SMCs) was analyzed using MTT assay and Transwell migration assay, respectively. The levels of contractile marker SM22α and synthetic marker osteopontin were measured by immunohistochemistry and Western blot to assess the phenotypic transition. RESULTS: The human varicose veins showed thickened intima, media and adventitia layers, increased synthetic VSMCs, as well as upregulated FOXC2-AS1 and FOXC2 expression. In vitro assays showed that FOXC2-AS1 overexpression promoted phenotypic transition, proliferation, and migration of SV-SMCs. However, the effect of FOXC2-AS1 overexpression could be abrogated by both FOXC2 silencing and the Notch signaling inhibitor FLI-06. Furthermore, FOXC2-AS1 overexpression activated the Notch pathway by upregulating FOXC2. CONCLUSION: FOXC2-AS1 overexpression promotes phenotypic transition, proliferation, and migration of SV-SMCs, at least partially, by activating the FOXC2-Notch pathway.


Sujet(s)
Mouvement cellulaire/physiologie , Prolifération cellulaire/physiologie , Facteurs de transcription Forkhead/métabolisme , Myocytes du muscle lisse/métabolisme , Veine saphène/métabolisme , Cellules cultivées , Humains , Myocytes du muscle lisse/anatomopathologie , Phénotype , Veine saphène/anatomopathologie , Transduction du signal , Régulation positive
10.
Differentiation ; 109: 28-33, 2019.
Article de Anglais | MEDLINE | ID: mdl-31494396

RÉSUMÉ

Vascular smooth muscle cells (VSMC) are highly specialized, and exhibit a contractile phenotype when mature and fully differentiated, being responsible for vessel homeostasis and blood pressure control. In response to pro-atherogenic stimuli VSMC alter their state of differentiation, increase proliferation and migration, resulting in SMC phenotypes ranging from contractile to synthetic. This variability is observed in cell morphology and expression level of marker genes for differentiation status. There is growing evidence that bone morphogenetic protein (BMP) signaling is involved in vascular diseases, including atherosclerosis. Here, we evaluated in vitro the role of specific agonists/antagonists belonging to the BMP pathway on dedifferentiation of VSMC harvested during early stages of atherosclerosis. RESULTS: Comparing primary VSMC isolated from aortas of susceptible ApoE-/- animals fed 8 weeks of western diet with their littermate controls fed usual diet, we observed that recombinant BMP4 was able to reduce SM22-alpha and alpha actin gene expression indicating dedifferentiation was under way. Unexpectedly, treatment with recombinant Gremlin-1, a known BMP antagonist, also reduced 4-6.5 folds gene expression of SM22-alpha, alpha-actin and, calponin, exclusively in VSMC from ApoE-/- animals, independently on the diet consumed. CONCLUSION: Our data show that BMP4 is capable of modulating of SM22-alpha and alpha actin gene expression, indicative of cell dedifferentiation in VSMC. Additionally, we report for first time that Gremlin-1 acts independently of the BMP pathway and selectively on VSMC from susceptible animals, reducing the expression of all genes evaluated.


Sujet(s)
Athérosclérose/anatomopathologie , Dédifférenciation cellulaire , Régulation de l'expression des gènes , Protéines et peptides de signalisation intercellulaire/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Animaux , Athérosclérose/métabolisme , Protéine morphogénétique osseuse de type 2/génétique , Protéine morphogénétique osseuse de type 2/métabolisme , Protéine morphogénétique osseuse de type 4/génétique , Protéine morphogénétique osseuse de type 4/métabolisme , Cellules cultivées , Protéines et peptides de signalisation intercellulaire/génétique , Souris , Souris de lignée C57BL , Souris invalidées pour les gènes ApoE , Protéines du muscle/génétique , Protéines du muscle/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Phénotype , Transduction du signal
11.
Int J Impot Res ; 31(2): 126-131, 2019 Mar.
Article de Anglais | MEDLINE | ID: mdl-30327570

RÉSUMÉ

Erectile dysfunction is a common condition that affects men over age 40. It is highly related to obesity. The corpus cavernosum is the most important structure involved in erection. The aim of this study was to evaluate the structure of the corpus cavernosum of mice fed with a high energy density diet (HED). At 3 months of age, male C57BL/6 mice were fed with a HED diet (50% lipids) or standard chow (SC) diet (10% lipids) for 14 weeks. Afterwards, the animals were euthanized and the corpus cavernosum was analyzed through stereology. Statistical significance was calculated by the student's t-test (p < 0.05). The group fed with HED diet showed higher values of body weight, blood pressure and higher rates of cholesterol, triglycerides, and glucose from the second week to the end of the experiment. The HED group showed a significant increase in the connective tissue (15%) and a decrease in smooth muscle fibers (41%). The testosterone concentration in the HED group was 63% lower than in SC animals. Animals fed with a HED presented reduced testosterone serum levels and morphological changes on the corpus cavernosum, which may be related to erectile dysfunction.


Sujet(s)
Matières grasses alimentaires/effets indésirables , Myocytes du muscle lisse/anatomopathologie , Pénis/anatomopathologie , Testostérone/sang , Animaux , Modèles animaux de maladie humaine , Dysfonctionnement érectile/étiologie , Mâle , Souris , Souris de lignée C57BL , Obésité/physiopathologie , Pénis/physiopathologie
12.
Life Sci ; 216: 279-286, 2019 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-30447304

RÉSUMÉ

AIMS: Inflammation is involved in diabetes-related vascular dysfunction. Estrogen receptor ESR2/ERß induces the expression of inducible nitric oxide (NO) synthase (iNOS) and inflammation. The present study investigated the effect of alloxan-induced type 1 diabetes on the iNOS and ESR2 expression and the effect of the chronic iNOS inhibition on the vascular smooth muscle dysfunction in diabetic female rats. In addition, we evaluated the involvement of ESR2 in iNOS expression. MAIN METHODS: Alloxan-induced diabetic female rats were treated or not with iNOS inhibitor (L-NIL). iNOS and ESR2 immunostaining, S-nitrosylated proteins and IL-1ß protein expression in aorta and plasmatic NO levels were analyzed. Contractile response to noradrenaline was analyzed in endothelium-denuded aorta. iNOS mRNA expression was analyzed in isolated aortic smooth muscle cells (ASMCs) of female rats, incubated with 22 mM glucose and an ESR2 antagonist. KEY FINDINGS: Aortic iNOS and ESR2 immunostaining, S-nitrosylated proteins, IL-1ß protein expression and plasmatic NO levels were all increased, whereas noradrenaline-induced contraction was reduced in aorta of diabetic female rats. With the exception of iNOS and ESR2 immunostaining, all these parameters were corrected by L-NIL treatment. High glucose increased iNOS mRNA expression in ASMCs, which was reduced by an ESR2 antagonist. SIGNIFICANCE: We demonstrated that increased iNOS-NO contributed to the impairment of the contractile response of aortic smooth muscle cells in female type 1 diabetic rats and that increased expression of iNOS may involve the participation of ESR2/ERß.


Sujet(s)
Diabète expérimental/physiopathologie , Diabète de type 1/physiopathologie , Récepteur bêta des oestrogènes/génétique , Inflammation/anatomopathologie , Nitric oxide synthase type II/génétique , Alloxane , Animaux , Aorte/anatomopathologie , Diabète expérimental/génétique , Diabète de type 1/génétique , Endothélium vasculaire/anatomopathologie , Femelle , Inflammation/génétique , Interleukine-1 bêta/génétique , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Monoxyde d'azote/métabolisme , Rats , Rat Wistar
13.
Biol. Res ; 52: 59, 2019. graf
Article de Anglais | LILACS | ID: biblio-1100911

RÉSUMÉ

OBJECTIVES: In varicose veins, vascular smooth muscle cells (VSMCs) often shows phenotypic transition and abnormal proliferation and migration. Evidence suggests the FOXC2-Notch pathway may be involved in the pathogenesis of varicose veins. Here, this study aimed to explore the role of long non-coding RNA FOXC2-AS1 (FOXC2 antisense RNA 1) in phenotypic transition, proliferation, and migration of varicose vein-derived VSMCs and to explore whether the FOXC2-Notch pathway was involved in this process. METHODS: The effect of FOXC2-AS1 on the proliferation and migration of human great saphenous vein smooth muscle cells (SV-SMCs) was analyzed using MTT assay and Transwell migration assay, respectively. The levels of contractile marker SM22α and synthetic marker osteopontin were measured by immunohistochemistry and Western blot to assess the phenotypic transition. RESULTS: The human varicose veins showed thickened intima, media and adventitia layers, increased synthetic VSMCs, as well as upregulated FOXC2-AS1 and FOXC2 expression. In vitro assays showed that FOXC2-AS1 overexpression promoted phenotypic transition, proliferation, and migration of SV-SMCs. However, the effect of FOXC2-AS1 overexpression could be abrogated by both FOXC2 silencing and the Notch signaling inhibitor FLI-06. Furthermore, FOXC2-AS1 overexpression activated the Notch pathway by upregulating FOXC2. CONCLUSION: FOXC2-AS1 overexpression promotes phenotypic transition, proliferation, and migration of SV-SMCs, at least partially, by activating the FOXC2-Notch pathway.


Sujet(s)
Humains , Veine saphène/métabolisme , Mouvement cellulaire/physiologie , Myocytes du muscle lisse/métabolisme , Prolifération cellulaire/physiologie , Facteurs de transcription Forkhead/métabolisme , Phénotype , Veine saphène/anatomopathologie , Transduction du signal , Régulation positive , Cellules cultivées , Myocytes du muscle lisse/anatomopathologie
14.
Asian J Androl ; 20(5): 505-510, 2018.
Article de Anglais | MEDLINE | ID: mdl-29893293

RÉSUMÉ

Erectile dysfunction is a common side effect of finasteride and dutasteride treatments. The objective of this study was to investigate the structural changes in the penis using a benign prostatic hyperplasia (BPH) rodent model treated with dutasteride or finasteride. Sixty male rats were divided into the following groups: C, untreated control rats; C + D, control rats receiving dutasteride; C + F, control rats receiving finasteride; H, untreated spontaneously hypertensive rats (SHRs); H + D, SHRs treated with dutasteride; and H + F, SHRs treated with finasteride. Treatments were performed for 40 days, and penises were collected immediately thereafter. The organs were analyzed using histomorphometric methods to determine the cross-sectional penile area, as well as the surface density (Sv) of smooth muscle fibers, connective tissue, elastic system fibers, and sinusoidal spaces of the corpus cavernosum. The results were compared using a one-way ANOVA with Bonferroni's posttest. Groups C + D and C + F had a significantly smaller penile cross-sectional area, but more elastic system fiber Sv compared to Group C. Group C + D showed less smooth muscle Sv, and Group H showed more connective tissue but a smaller sinusoidal space Sv in the corpus cavernosum compared to Group C. Groups H + D and H + F had less smooth muscle Sv than Group H. Group H + D also had more connective tissue and elastic system fiber Sv than Group H. Both dutasteride and finasteride promoted penile modifications in the control rat penis, although this affect was greater in Group H animals. In this rodent model, dutasteride was the drug that most affected the corpus cavernosum.


Sujet(s)
Inhibiteurs de la 5-alpha réductase/pharmacologie , Dutastéride/pharmacologie , Finastéride/pharmacologie , Muscles lisses/effets des médicaments et des substances chimiques , Pénis/effets des médicaments et des substances chimiques , Hyperplasie de la prostate/anatomopathologie , Inhibiteurs de la 5-alpha réductase/usage thérapeutique , Animaux , Modèles animaux de maladie humaine , Dutastéride/usage thérapeutique , Finastéride/usage thérapeutique , Mâle , Muscles lisses/anatomopathologie , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/anatomopathologie , Pénis/anatomopathologie , Prostate/effets des médicaments et des substances chimiques , Prostate/anatomopathologie , Hyperplasie de la prostate/traitement médicamenteux , Rats
15.
PLoS One ; 13(5): e0197210, 2018.
Article de Anglais | MEDLINE | ID: mdl-29750813

RÉSUMÉ

Vascular smooth muscle cells (VSMC) dedifferentiation from a contractile to a synthetic phenotype contributes to atherosclerosis. Atherosclerotic tissue has a chronic inflammatory component with high levels of tumor necrosis factor-α (TNF-α). VSMC of atheromatous plaques have increased autophagy, a mechanism responsible for protein and intracellular organelle degradation. The aim of this study was to evaluate whether TNF-α induces phenotype switching of VSMCs and whether this effect depends on autophagy. Rat aortic Vascular smooth A7r5 cell line was used as a model to examine the phenotype switching and autophagy. These cells were stimulated with TNF-α 100 ng/mL. Autophagy was determined by measuring LC3-II and p62 protein levels. Autophagy was inhibited using chloroquine and siRNA Beclin1. Cell dedifferentiation was evaluated by measuring the expression of contractile proteins α-SMA and SM22, extracellular matrix protein osteopontin and type I collagen levels. Cell proliferation was measured by [3H]-thymidine incorporation and MTT assay, and migration was evaluated by wound healing and transwell assays. Expression of IL-1ß, IL-6 and IL-10 was assessed by ELISA. TNF-α induced autophagy as determined by increased LC3-II (1.91±0.21, p<0.001) and decreased p62 (0.86±0.02, p<0.05) when compared to control. Additionally, TNF-α decreased α-SMA (0.74±0.12, p<0.05) and SM22 (0.54±0.01, p<0.01) protein levels. Consequently, TNF-α induced migration (1.25±0.05, p<0.05), proliferation (2.33±0.24, p<0.05), and the secretion of IL-6 (258±53, p<0.01), type I collagen (3.09±0.85, p<0.01) and osteopontin (2.32±0.46, p<0.01). Inhibition of autophagy prevented all the TNF-α-induced phenotypic changes. TNF-α induces phenotype switching in A7r5 cell line by a mechanism that required autophagy. Therefore, autophagy may be a potential therapeutic target for the treatment of atherosclerosis.


Sujet(s)
Athérosclérose/métabolisme , Autophagie , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Animaux , Athérosclérose/anatomopathologie , Lignée cellulaire , Interleukine-10/métabolisme , Interleukine-1 bêta/métabolisme , Interleukine-6/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Rats
16.
Oxid Med Cell Longev ; 2018: 5835072, 2018.
Article de Anglais | MEDLINE | ID: mdl-30671170

RÉSUMÉ

In metabolic diseases, the increased reactive oxygen species (ROS) represents one of the pathogenic mechanisms for vascular disease probably by promoting vascular smooth muscle cell (SMC) proliferation that contributes to the development of arterial remodeling and stenosis, hypertension, and atherosclerosis. Therefore, this work was undertaken to evaluate the participation of ROS from NADPH oxidase and mitochondria in the proliferation of SMCs from the aorta in a model of metabolic syndrome induced by sucrose feeding in rats. After 24 weeks, sucrose-fed (SF) rats develop hypertension, intra-abdominal obesity, hyperinsulinemia, and hyperleptinemia. In addition SMCs from SF rats had a higher growth rate and produce more ROS than control cells. The treatment of SMCs with DPI and apocynin to inhibit NADPH oxidase and with tempol to scavenge superoxide anion significantly blocked the proliferation of both SF and control cells suggesting the participation of NADPH oxidase as a source of superoxide anion. MitoTEMPO, which targets mitochondria within the cell, also significantly inhibited the proliferation of SMCs having a greater effect on cells from SF than from the control aorta. The higher rate of cell growth from the SF aorta is supported by the increased content of cyclophilin A and CD147, proteins involved in the mechanism of cell proliferation. In addition, caldesmon, α-actin, and phosphorylated myosin light chain, contractile phenotype proteins, were found significantly lower in SF cells in no confluent state and increased in confluent state but without difference between both cell types. Our results suggest that ROS from NADPH oxidase and mitochondria significantly participate in the difference found in the rate of cell growth between SF and control cells.


Sujet(s)
Aorte/enzymologie , Prolifération cellulaire , Syndrome métabolique X/enzymologie , Mitochondries du muscle/enzymologie , Muscles lisses vasculaires/enzymologie , Myocytes du muscle lisse/enzymologie , NADPH oxidase/métabolisme , Espèces réactives de l'oxygène/métabolisme , Animaux , Aorte/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Syndrome métabolique X/anatomopathologie , Mitochondries du muscle/anatomopathologie , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Rats , Rat Wistar
17.
Neurourol Urodyn ; 37(1): 148-152, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-28598513

RÉSUMÉ

AIMS: To study the bladder structure of fetuses with prune belly syndrome (PBS). METHODS: We studied three bladders obtained from three male fetuses with PBS and seven bladders from seven male fetuses without anomalies. Each bladder was dissected and embedded in paraffin, from which 5 µm thick sections were obtained and stained with Masson's trichrome (to quantify connective tissue and smooth muscle) and picrosirius red with polarization (to observe collagen). Immunohistochemistry with tubulin (Tubulin, beta III, Mouse Monoclonal Antibody) was applied to observe the bladder nerves. The images were captured with an Olympus BX51 microscope and Olympus DP70 camera. The stereological analysis was done with the Image Pro and Image J programs, using a grid to determine volumetric densities (Vv). Means were statistically compared using the Mann-Whitney test (P < 0.05). RESULTS: Quantitative analysis documented that smooth muscle fibers were significantly smaller (P = 0.04) in PBS fetuses (9.67% to 17.75%, mean = 13.2%) compared to control group (13.33% to 26.56%, mean = 17.43%). The analysis of collagen fibers showed predominance of green in the control group, suggesting collagen type III presence, and predominance of red in the in PBS fetal bladders, suggesting collagen type I presence in this group. The qualitative analysis of the nerves with immunohistochemistry with tubulin showed predominance of nerves in the control group. CONCLUSION: The bladder in PBS had lower concentrations of smooth muscle fibers, collagen type III, and nerves. These structural alterations can be one of the factors involved in urinary tract abnormality such as distended bladder in patients with PBS.


Sujet(s)
Muscles lisses/anatomopathologie , Syndrome de Prune Belly/anatomopathologie , Vessie urinaire/anatomopathologie , Collagène de type III/métabolisme , Foetus , Humains , Immunohistochimie , Mâle , Muscles lisses/métabolisme , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Syndrome de Prune Belly/métabolisme , Vessie urinaire/métabolisme
18.
Stem Cell Res Ther ; 8(1): 220, 2017 10 03.
Article de Anglais | MEDLINE | ID: mdl-28974252

RÉSUMÉ

BACKGROUND: Experimental research has reported beneficial effects of mesenchymal stromal cell (MSC) therapy in pulmonary arterial hypertension (PAH). However, these studies either were based on prophylactic protocols or assessed basic remodeling features without evaluating possible mechanisms. We analyzed the effects of MSC therapy on lung vascular remodeling and hemodynamics and its possible mechanisms of action in monocrotaline (MCT)-induced PAH. METHODS: Twenty-eight Wistar rats were randomly divided into two groups. In the PAH group, animals received MCT 60 mg/kg intraperitoneally, while a control group received saline (SAL) instead. On day 14, both groups were further randomized to receive 105 adipose-derived MSCs or SAL intravenously (n = 7/group). On day 28, right ventricular systolic pressure (RVSP) and the gene expression of mediators associated with apoptosis, inflammation, fibrosis, Smad-1 levels, cell proliferation, and endothelial-mesenchymal transition were determined. In addition, lung histology (smooth muscle cell proliferation and plexiform-like injuries), CD68+ and CD163+ macrophages, and plasma levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) were evaluated. RESULTS: In the PAH group, adipose-derived MSCs, compared to SAL, reduced mean RVSP (29 ± 1 vs 39 ± 2 mmHg, p < 0.001), lung tissue collagen fiber content, smooth muscle cell proliferation, CD68+ macrophages, interleukin-6 expression, and the antiapoptotic mediators Bcl-2 and survivin. Conversely, expression of the proapoptotic mediator procaspase-3 and plasma VEGF increased, with no changes in PDGF. In the pulmonary artery, MSCs dampened the endothelial-mesenchymal transition. CONCLUSION: In MCT-induced PAH, MSC therapy reduced lung vascular remodeling, thus improving hemodynamics. These beneficial effects were associated with increased levels of proapoptotic markers, mesenchymal-to-endothelial transition, reduced cell proliferation markers, and inflammation due to a shift away from the M1 phenotype.


Sujet(s)
Tissu adipeux/cytologie , Hémodynamique/physiologie , Hypertension pulmonaire/thérapie , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/cytologie , Tissu adipeux/métabolisme , Animaux , Antigènes CD/génétique , Antigènes CD/métabolisme , Prolifération cellulaire , Collagène/génétique , Collagène/métabolisme , Régulation de l'expression des gènes , Hypertension pulmonaire/induit chimiquement , Hypertension pulmonaire/génétique , Hypertension pulmonaire/anatomopathologie , Interleukine-6/génétique , Interleukine-6/métabolisme , Poumon/vascularisation , Poumon/métabolisme , Poumon/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Mâle , Cellules souches mésenchymateuses/métabolisme , Protéines associées aux microtubules/génétique , Protéines associées aux microtubules/métabolisme , Monocrotaline , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Facteur de croissance dérivé des plaquettes/génétique , Facteur de croissance dérivé des plaquettes/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Rats , Rat Wistar , Protéine Smad-1/génétique , Protéine Smad-1/métabolisme , Survivine , Facteur de croissance endothéliale vasculaire de type A/génétique , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Remodelage vasculaire/génétique
19.
Adv Exp Med Biol ; 998: 151-160, 2017.
Article de Anglais | MEDLINE | ID: mdl-28936738

RÉSUMÉ

Vascular calcification is a tightly regulated process that increases during ageing and occurs mainly in patients with diabetes and chronic renal failure. Exosomes are small membrane vesicles that are synthesized in a particular population of endosomes, also called multivesicular bodies, by inside budding into the lumen of the compartment. After fusion of exosome with the plasma membrane, these internal vesicles are secreted. Exosomes have a defined set of membrane and cytosolic proteins. The physiological function of exosomes is still a matter of debate. Investigators implicated microvesicles/exosomes as a specific signaling mechanism to induce vascular mineralization during vascular smooth muscle cells phenotypic transition. Vascular wall from healthy individual exhibit exosomes loaded with calcification inhibitors such as Fetuin A and MGP. Conversely, calcifying conditions induce secretion of exosomes, characterized by decreased calcifying inhibitors and increased phosphatidyl serine and Annexin A6 content, which serves as a nidus for vascular calcification.


Sujet(s)
Signalisation calcique , Calcium/métabolisme , Exosomes/métabolisme , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Calcification vasculaire/métabolisme , Animaux , Artères/métabolisme , Artères/anatomopathologie , Exosomes/anatomopathologie , Humains , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Calcification vasculaire/anatomopathologie
20.
Atherosclerosis ; 265: 14-21, 2017 Oct.
Article de Anglais | MEDLINE | ID: mdl-28829997

RÉSUMÉ

BACKGROUND AND AIMS: Obesity and diabetes potentiate vascular calcification by increasing vascular smooth muscle cells osteoblastic differentiation mediated by the transcription factor Msx2 and bone morphogenetic protein-2 signaling. However, Bmp-2/Msx2 crosstalk to induce VSMC osteogenic phenotype transition and calcification is poorly understood in diabetes. We aimed to investigate mechanisms underlying Bmp-2-driven VSMC osteogenic differentiation and calcification in leptin-deficient ob/ob mice. METHODS: We incubated VSMC from ob/ob mice and wild type C57BL/6 littermates with or without Bmp-2. We used loss-of-function experiments to investigate the role of Msx2 in Bmp-2-induced ob/ob VSMC osteochondrogenic differentiation and calcification by transfecting Msx2 siRNA into VSMC. RESULTS: Baseline ob/ob VSMC and aorta showed increased Msx2, Runx2, alkaline phosphatase mRNA and protein expression, which further increased in Bmp-2-incubated ob/ob VSMC, therefore augmenting ob/ob VSMC calcification in comparison to wild type VSMC. Accordingly, signaling pathways to induce VSMC osteogenic differentiation, such as Smad1/5 phosphorylation increased in ob/ob versus wild type aorta. In comparison to wild type VSMC, Msx2 siRNA transfected VSMC decreased Bmp-2-dependent osteochondrogenic differentiation response by abrogating Msx2, Runx2, Alpl expression in ob/ob but not in wild type VSMC. Nonetheless, Msx2 inhibition did not decrease calcification in Bmp-2 stimulated ob/ob VSMC in vitro. CONCLUSIONS: Our data support a crucial role of Msx2 for ob/ob VSMC osteochondrogenic differentiation, however, Msx2 signaling alone is not sufficient for ob/ob VSMC calcification after Bmp-2 stimulation in vitro. These findings can be translated into novel perspectives for the understanding of the mechanisms and to provide therapeutic targets underlying vascular calcification in type 2 diabetes.


Sujet(s)
Maladies de l'aorte/métabolisme , Protéines à homéodomaine/métabolisme , Insulinorésistance , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/métabolisme , Obésité/métabolisme , Ostéoblastes/métabolisme , Ostéogenèse , Calcification vasculaire/métabolisme , Animaux , Aorte/métabolisme , Aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/anatomopathologie , Protéine morphogénétique osseuse de type 2/déficit , Protéine morphogénétique osseuse de type 2/génétique , Cellules cultivées , Modèles animaux de maladie humaine , Prédisposition génétique à une maladie , Protéines à homéodomaine/génétique , Insulinorésistance/génétique , Mâle , Souris de lignée C57BL , Souris knockout , Souris obèse , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Obésité/génétique , Obésité/anatomopathologie , Ostéoblastes/anatomopathologie , Phénotype , Interférence par ARN , Transduction du signal , Transfection , Calcification vasculaire/génétique , Calcification vasculaire/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE