Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
1.
Int J Biol Sci ; 17(10): 2399-2416, 2021.
Article de Anglais | MEDLINE | ID: mdl-34326683

RÉSUMÉ

Myocardial aging increases the cardiovascular risk in the elderly. The Receptor for Advanced Glycation End-products (RAGE) is involved in age-related disorders. The soluble isoform (sRAGE) acts as a scavenger blocking the membrane-bound receptor activation. This study aims at investigating RAGE contribution to age-related cardiac remodeling. We analyzed the cardiac function of three different age groups of female Rage-/- and C57BL/6N (WT) mice: 2.5- (Young), 12- (Middle-age, MA) and 21-months (Old) old. While aging, Rage-/- mice displayed an increase in left ventricle (LV) dimensions compared to age-matched WT animals, with the main differences observed in the MA groups. Rage-/- mice showed higher fibrosis and a larger number of α-Smooth Muscle Actin (SMA)+ cells with age, along with increased expression of pro-fibrotic Transforming Growth Factor (TGF)-ß1 pathway components. RAGE isoforms were undetectable in LV of WT mice, nevertheless, circulating sRAGE declined with aging and inversely associated with LV diastolic dimensions. Human cardiac fibroblasts stimulated with sRAGE exhibited a reduction in proliferation, pro-fibrotic proteins and TGF-beta Receptor 1 (TGFbR1) expression and Smad2-3 activation. Finally, sRAGE administration to MA WT animals reduced cardiac fibrosis. Hence, our work shows that RAGE associates with age-dependent myocardial changes and indicates sRAGE as an inhibitor of cardiac fibroblasts differentiation and age-dependent cardiac fibrosis.


Sujet(s)
Actines/métabolisme , Vieillissement , Myocarde/métabolisme , Récepteur spécifique des produits finaux de glycosylation avancée/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Lignée cellulaire , Femelle , Fibroblastes/métabolisme , Fibrose , Humains , Souris , Souris de lignée C57BL , Myocarde/anatomopathologie , Isoformes de protéines/métabolisme
2.
Cells ; 9(1)2020 01 08.
Article de Anglais | MEDLINE | ID: mdl-31936351

RÉSUMÉ

: Background: Marfan syndrome (MFS) is a genetic disease, characterized by thoracic aortic aneurysm (TAA), which treatment is to date purely surgical. Understanding of novel molecular targets is mandatory to unveil effective pharmacological approaches. Cyclophilin A (CyPA) and its receptor EMMPRIN are associated with several cardiovascular diseases, including abdominal aortic aneurysm. Here, we envisioned the contribution of CyPA/EMMPRIN axis in MFS-related TAA. METHODS: We obtained thoracic aortic samples from healthy controls (HC) and MFS patients' aortas and then isolated vascular smooth muscle cells (VSMC) from the aortic wall. RESULTS: our findings revealed that MFS aortic tissue samples isolated from the dilated zone of aorta showed higher expression levels of EMMPRIN vs. MFS non-dilated aorta and HC. Interestingly, angiotensin II significantly stimulated CyPA secretion in MFS-derived VSMC (MFS-VSMC). CyPA treatment on MFS-VSMC led to increased levels of EMMPRIN and other MFS-associated pro-fibrotic mediators, such as TGF-ß1 and collagen I. These molecules were downregulated by in vitro treatment with CyPA inhibitor MM284. Our results suggest that CyPA/EMMPRIN axis is involved in MFS-related TAA development, since EMMPRIN is upregulated in the dilated zone of MFS patients' TAA and the inhibition of its ligand, CyPA, downregulated EMMPRIN and MFS-related markers in MFS-VSMC. CONCLUSIONS: these insights suggest both a novel detrimental role for CyPA/EMMPRIN axis and its inhibition as a potential therapeutic strategy for MFS-related TAA treatment.


Sujet(s)
Anévrysme de l'aorte thoracique/anatomopathologie , Antigènes CD147/métabolisme , Cyclophiline A/métabolisme , Fibrose/anatomopathologie , Syndrome de Marfan/physiopathologie , Facteur de croissance transformant bêta-1/métabolisme , Anévrysme de l'aorte thoracique/métabolisme , Études cas-témoins , Fibrose/métabolisme , Humains
3.
Theranostics ; 9(8): 2224-2234, 2019.
Article de Anglais | MEDLINE | ID: mdl-31149040

RÉSUMÉ

Marfan syndrome (MFS) is a rare genetic disease characterized by a matrix metalloproteases (MMPs) dysregulation that leads to extracellular matrix degradation. Consequently, MFS patients are prone to develop progressive thoracic aortic enlargement and detrimental aneurysm. Since MMPs are activated by the extracellular MMP inducer (EMMPRIN) protein, we determined whether its plasmatic soluble form (sEMMPRIN) may be considered a marker of thoracic aortic ectasia (AE). Methods: We compared plasma sEMMPRIN levels of 42 adult Caucasian MFS patients not previously subjected to aortic surgery with those of matched healthy controls (HC) by ELISA. In the MFS cohort we prospectively evaluated the relationship between plasma sEMMPRIN levels and the main MFS-related manifestations. Results: MFS patients had lower plasma sEMMPRIN levels (mean±SD: 2071±637 pg/ml) than HC (2441±642 pg/ml, p=0.009). Amongst all considered MFS-related clinical features, we found that only aortic root dilatation associated with circulating sEMMPRIN levels. Specifically, plasma sEMMPRIN levels negatively correlated with aortic Z-score (r=-0.431, p=0.004), and were significantly lower in patients with AE (Z-score≥2, 1788±510 pg/ml) compared to those without AE (Z-score<2, 2355±634 pg/ml; p=0.003). ROC curve analysis revealed that plasma sEMMPRIN levels discriminated patients with AE (AUC [95%CI]: 0.763 [0.610-0.916], p=0.003) with 85.7% sensitivity, 76.2% specificity, and 81% accuracy. We defined plasma sEMMPRIN levels ≤2246 pg/ml as the best threshold discriminating the presence of AE in MFS patients with an odds ratio [95%CI] of 19.2 [3.947-93.389] (p<0.001). Conclusions: MFS patients are characterized by lower sEMMPRIN levels than HC. Notably, plasma sEMMPRIN levels are strongly associated with thoracic AE.


Sujet(s)
Aorte/anatomopathologie , Antigènes CD147/sang , Syndrome de Marfan/diagnostic , Adulte , Marqueurs biologiques/sang , Dilatation pathologique/sang , Dilatation pathologique/anatomopathologie , Femelle , Humains , Mâle , Syndrome de Marfan/sang , Sensibilité et spécificité
4.
Int J Mol Sci ; 20(10)2019 May 15.
Article de Anglais | MEDLINE | ID: mdl-31096574

RÉSUMÉ

Arrhythmogenic cardiomyopathy (ACM) is a genetic disorder characterized by the progressive substitution of functional myocardium with noncontractile fibro-fatty tissue contributing to ventricular arrhythmias and sudden cardiac death. Cyclophilin A (CyPA) is a ubiquitous protein involved in several pathological mechanisms, which also characterize ACM (i.e., fibrosis, inflammation, and adipogenesis). Nevertheless, the involvement of CyPA in ACM cardiac remodeling has not been investigated yet. Thus, we first evaluated CyPA expression levels in the right ventricle (RV) tissue specimens obtained from ACM patients and healthy controls (HC) by immunohistochemistry. Then, we took advantage of ACM- and HC-derived cardiac mesenchymal stromal cells (C-MSC) to assess CyPA modulation during adipogenic differentiation. Interestingly, CyPA was more expressed in the RV sections obtained from ACM vs. HC subjects and positively correlated with the adipose replacement extent. Moreover, CyPA was upregulated at early stages of C-MSC adipogenic differentiation and was secreted at higher level over time in ACM- derived C-MSC. Our study provides novel ex vivo and in vitro information on CyPA expression in ACM remodeling paving the way for future C-MSC-based mechanistic and therapeutic investigations.


Sujet(s)
Troubles du rythme cardiaque/métabolisme , Cardiomyopathies/métabolisme , Cyclophiline A/métabolisme , Remodelage ventriculaire , Adipogenèse/physiologie , Tissu adipeux/anatomopathologie , Troubles du rythme cardiaque/anatomopathologie , Cardiomyopathies/anatomopathologie , Différenciation cellulaire , Cyclophiline A/génétique , Mort subite cardiaque/anatomopathologie , Fibrose , Expression des gènes , Ventricules cardiaques/métabolisme , Ventricules cardiaques/anatomopathologie , Humains , Inflammation , Cellules souches mésenchymateuses/anatomopathologie , Myocarde
5.
Am J Pathol ; 189(2): 339-353, 2019 02.
Article de Anglais | MEDLINE | ID: mdl-30448404

RÉSUMÉ

Patients affected by Duchenne muscular dystrophy (DMD) develop a progressive dilated cardiomyopathy characterized by inflammatory cell infiltration, necrosis, and cardiac fibrosis. Standard treatments consider the use of ß-blockers and angiotensin-converting enzyme inhibitors that are symptomatic and unspecific toward DMD disease. Medications that target DMD cardiac fibrosis are in the early stages of development. We found immunoproteasome dysregulation in affected hearts of mdx mice (murine animal model of DMD) and cardiomyocytes derived from induced pluripotent stem cells of patients with DMD. Interestingly, immunoproteasome inhibition ameliorated cardiomyopathy in mdx mice and reduced the development of cardiac fibrosis. Establishing the immunoproteasome inhibition-dependent cardioprotective role suggests the possibility of modulating the immunoproteasome as new and clinically relevant treatment to rescue dilated cardiomyopathy in patients with DMD.


Sujet(s)
Cardiomyopathies , Myopathie de Duchenne , Myocytes cardiaques , Proteasome endopeptidase complex/immunologie , Animaux , Cardiomyopathies/immunologie , Cardiomyopathies/anatomopathologie , Fibrose , Humains , Cellules souches pluripotentes induites/immunologie , Cellules souches pluripotentes induites/anatomopathologie , Mâle , Souris , Souris de lignée mdx , Myopathie de Duchenne/immunologie , Myopathie de Duchenne/anatomopathologie , Myocytes cardiaques/immunologie , Myocytes cardiaques/anatomopathologie
6.
J Transl Med ; 16(1): 352, 2018 12 12.
Article de Anglais | MEDLINE | ID: mdl-30541573

RÉSUMÉ

BACKGROUND: To date the TGF-ß1 activation mediated by integrin ανß5 during fibrosis is well-known. This process has been shown also in the heart, where cardiac fibroblasts (CF) differentiate into α-smooth muscle actin (α-SMA)-positive myofibroblasts (MyoFB). Here, we studied the effects on CF, isolated by spontaneously hypertensive rats (SHR), of integrin ανß5 inhibition in MyoFB differentiation. METHODS: Staining and immunohistochemistry were performed on rat cardiac tissue. CF were isolated by enzymatic digestion from SHR (SHR-CF) and normotensive WKY (WKY-CF) rat hearts and then treated for in vitro evaluation. RESULTS: SHR heart tissues revealed a higher TGF-ß1 expression vs. WKY samples. SHR-CF showed an enhanced SMAD2/3 activation and an up-regulated expression of α-SMA, a typical MyoFB marker, especially after TGF-ß1 treatment. Immunostaining on cardiac tissues revealed a higher expression of integrin ανß5 in SHR vs. WKY rat hearts. In vitro results confirmed the up-regulation of integrin ανß5 expression in SHR-CF at basal condition and after TGF-ß1 treatment, in comparison with WKY-CF. Inhibition of integrin ανß5 by cilengitide treatment led a decreased expression of ανß5, collagen I, and α-SMA in SHR-CF vs. WKY-CF, resulting in a diminished differentiation of CF into MyoFB. Taking together, results suggested that SHR-CF are more susceptible to TGF-ß1, showing an up-regulated activation of SMAD2/3 signaling, and an increased ανß5, α-SMA, and collagen I expression. Hypertension stimulus promoted an up-regulation of integrin ανß5 on SHR cardiac tissue and its in vitro inhibition reverted pro-fibrotic events of SHR-CF. CONCLUSION: Inhibition of integrin ανß5 exerted by cilengitide strongly diminished SHR-CF differentiation into detrimental MyoFB. So, integrin ανß5 might be considered a novel therapeutic target and cilengitide an effective pharmacological tool to limit the progression of hypertension-induced cardiac fibrosis.


Sujet(s)
Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Myocarde/métabolisme , Myocarde/anatomopathologie , Récepteur vitronectine/antagonistes et inhibiteurs , Actines/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Pression sanguine/effets des médicaments et des substances chimiques , Collagène de type I/métabolisme , Diastole/effets des médicaments et des substances chimiques , Mâle , Myofibroblastes/effets des médicaments et des substances chimiques , Myofibroblastes/métabolisme , Myofibroblastes/anatomopathologie , Rats de lignée SHR , Rats de lignée WKY , Récepteur vitronectine/génétique , Récepteur vitronectine/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéines Smad/métabolisme , Venins de serpent/pharmacologie , Systole/effets des médicaments et des substances chimiques , Facteur de croissance transformant bêta-1/métabolisme , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/génétique
7.
Prog Cardiovasc Dis ; 61(3-4): 328-335, 2018.
Article de Anglais | MEDLINE | ID: mdl-30041021

RÉSUMÉ

Marfan Syndrome (MFS) is a rare connective tissue disorder, resulting from mutations in the fibrillin-1 gene, characterized by pathologic phenotypes in multiple organs, the most detrimental of which affects the thoracic aorta. Indeed, thoracic aortic aneurysms (TAA), leading to acute dissection and rupture, are today the major cause of morbidity and mortality in adult MFS patients. Therefore, there is a compelling need for novel therapeutic strategies to delay TAA progression and counteract aortic dissection occurrence. Unfortunately, the wide phenotypic variability of MFS patients, together with the lack of a complete genotype-phenotype correlation, have represented until now a barrier hampering the conduction of translational studies aimed to predict disease prognosis and drug discovery. In this review, we will illustrate available therapeutic strategies to improve the health of MFS patients. Starting from gold standard surgical overtures and the description of the main pharmacological approaches, we will comprehensively review the state-of-the-art of in vivo MFS models and discuss recent clinical pharmacogenetic results. Finally, we will focus on induced pluripotent stem cells (iPSC) as a technology that, if integrated with preclinical research and pharmacogenetics, could contribute in determining the best therapeutic approach for each MFS patient on the base of individual differences. Finally, we will suggest the integration of preclinical studies, pharmacogenetics and iPSC technology as the most likely strategy to help solve the composite puzzle of precise medicine in this condition.


Sujet(s)
Anévrysme de l'aorte thoracique , , Syndrome de Marfan , Adulte , /étiologie , /mortalité , /prévention et contrôle , Anévrysme de l'aorte thoracique/étiologie , Anévrysme de l'aorte thoracique/mortalité , Humains , Syndrome de Marfan/complications , Syndrome de Marfan/génétique , Syndrome de Marfan/thérapie , Test pharmacogénomique , Médecine de précision/méthodes , Pronostic
8.
Stem Cell Res ; 28: 21-24, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29414413

RÉSUMÉ

Becker muscular dystrophy (BMD) is a dystrophinopathy caused by mutations in the dystrophin gene on chromosome Xp21. BMD mutations result in truncated semi-functional dystrophin isoforms. Consequently, less severe clinical symptoms become apparent later in life compared to Duchenne muscular dystrophy. Dermal fibroblasts from a BMD patient were electroporated with episomal plasmids containing reprogramming factors to create the induced pluripotent stem cell line: CCMi002BMD-A-9 that showed pluripotent markers, were karyotypically normal and capable of trilineage differentiation. MLPA analyses performed on DNA extracted from CCMi002BMD-A-9 showed an in-frame deletion of exons 45 to 55 (CCMi002BMD-A-9 Δ45-55).


Sujet(s)
Techniques de culture cellulaire/méthodes , Dystrophine/génétique , Exons/génétique , Cellules souches pluripotentes induites/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , Délétion de séquence/génétique , Adulte , Humains , Mâle
9.
Pharmacol Res ; 127: 77-91, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-28235633

RÉSUMÉ

Over the past two decades cardiac cell therapy (CCT) has emerged as a promising new strategy to cure heart diseases at high unmet need. Thousands of patients have entered clinical trials for acute or chronic heart conditions testing different cell types, including autologous or allogeneic bone marrow (BM)-derived mononuclear or selected cells, BM- or adipose tissue-derived mesenchymal cells, or cardiac resident progenitors based on their potential ability to regenerate scarred or dysfunctional myocardium. Nowadays, the original enthusiasm surrounding the regenerative medicine field has been cushioned by a cumulative body of evidence indicating an inefficient or modest efficacy of CCT in improving cardiac function, along with the continued lack of indisputable proof for long-term prognostic benefit. In this review, we have firstly comprehensively outlined the positive and negative results of cell therapy studies in patients with acute myocardial infarction, refractory angina and chronic heart failure. Next, we have discussed cell therapy- and patient-related variables (e.g. cell intrinsic and extrinsic characteristics as well as criteria of patient selection and proposed methodologies) that might have dampened the efficacy of past cell therapy trials. Finally, we have addressed critical factors to be considered before embarking on further clinical trials.


Sujet(s)
Angine de poitrine/thérapie , Thérapie cellulaire et tissulaire/méthodes , Défaillance cardiaque/thérapie , Infarctus du myocarde/thérapie , Humains , Sélection de patients
10.
Stem Cell Res ; 25: 128-131, 2017 12.
Article de Anglais | MEDLINE | ID: mdl-29127875

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is caused by abnormalities in the dystrophin gene and is clinically characterised by childhood muscle degeneration and cardiomyopathy. We produced an induced pluripotent stem cell line from a DMD patient's dermal fibroblasts by electroporation with episomal vectors containing: hL-MYC, hLIN28, hSOX2, hKLF4, hOCT3/4. The resultant DMD iPSC line (CCMi001DMD-A-3) displayed iPSC morphology, expressed pluripotency markers, possessed trilineage differentiation potential and was karyotypically normal. MLPA analyses performed on DNA extracted from CCMi001DMD-A-3 showed a deletion of exons 49 and 50 (CCMi001DMD-A-3, ∆49, ∆50).


Sujet(s)
Exons/génétique , Cellules souches pluripotentes induites/cytologie , Myopathie de Duchenne/enzymologie , Adulte , Cellules cultivées , Reprogrammation cellulaire/génétique , Humains , Cellules souches pluripotentes induites/métabolisme , Mâle
11.
Stem Cells Int ; 2017: 5648690, 2017.
Article de Anglais | MEDLINE | ID: mdl-29375624

RÉSUMÉ

Cardiac cell-based therapy has emerged as a novel therapeutic option for patients dealing with untreatable refractory angina (RA). However, after more than a decade of controlled studies, no definitive consensus has been reached regarding clinical efficacy. Although positive results in terms of surrogate endpoints have been suggested by early and phase II clinical studies as well as by meta-analyses, the more recent reports lacked the provision of definitive response in terms of hard clinical endpoints. Regrettably, pivotal trials designed to conclusively determine the efficacy of cell-based therapeutics in such a challenging clinical condition are therefore still missing. Considering this, a comprehensive reappraisal of cardiac cell-based therapy role in RA seems warranted and timely, since a number of crucial cell- and patient-related aspects need to be systematically analysed. As an example, the large variability in efficacy endpoint selection appears to be a limiting factor for the advancement of cardiac cell-based therapy in the field. This review will provide an overview of the key elements that may have influenced the results of cell-based trials in the context of RA, focusing in particular on the understanding at which the extent of angina-related endpoints may predict cell-based therapeutic efficacy.

12.
Cell Mol Life Sci ; 74(2): 267-277, 2017 01.
Article de Anglais | MEDLINE | ID: mdl-27535662

RÉSUMÉ

Marfan syndrome (MFS) is a connective tissue disorder with multiple organ manifestations. The genetic cause of this syndrome is the mutation of the FBN1 gene, encoding the extracellular matrix (ECM) protein fibrillin-1. This genetic alteration leads to the degeneration of microfibril structures and ECM integrity in the tunica media of the aorta. Indeed, thoracic aortic aneurysm and dissection represent the leading cause of death in MFS patients. To date, the most effective treatment option for this pathology is the surgical substitution of the damaged aorta. To highlight novel therapeutic targets, we review the molecular mechanisms related to MFS etiology in vascular smooth muscle cells, the foremost cellular type involved in MFS pathogenesis.


Sujet(s)
Anévrysme/complications , Anévrysme/anatomopathologie , Aorte/anatomopathologie , Syndrome de Marfan/complications , Syndrome de Marfan/anatomopathologie , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/anatomopathologie , Animaux , Humains , Mécanotransduction cellulaire
13.
J Cell Mol Med ; 21(5): 871-880, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-27860185

RÉSUMÉ

The recruitment of bone marrow (BM)-derived progenitor cells to the lung is related to pulmonary remodelling and the pathogenesis of pulmonary hypertension (PH). Although sildenafil is a known target in PH treatment, the underlying molecular mechanism is still elusive. To test the hypothesis that the therapeutic effect of sildenafil is linked to the reduced recruitment of BM-derived progenitor cells, we induced pulmonary remodelling in rats by two-week exposure to chronic hypoxia (CH, 10% oxygen), a trigger of BM-derived progenitor cells. Rats were treated with either placebo (saline) or sildenafil (1.4 mg/kg/day ip) during CH. Control rats were kept in room air (21% oxygen) with no treatment. As expected, sildenafil attenuated the CH-induced increase in right ventricular systolic pressure and right ventricular hypertrophy. However, sildenafil suppressed the CH-induced increase in c-kit+ cells in the adventitia of pulmonary arteries. Moreover, sildenafil reduced the number of c-kit+ cells that colocalize with tyrosine kinase receptor 2 (VEGF-R2) and CD68 (a marker for macrophages), indicating a positive effect on moderating hypoxia-induced smooth muscle cell proliferation and inflammation without affecting the pulmonary levels of hypoxia-inducible factor (HIF)-1α. Furthermore, sildenafil depressed the number of CXCR4+ cells. Collectively, these findings indicate that the improvement in pulmonary haemodynamic by sildenafil is linked to decreased recruitment of BM-derived c-kit+ cells in the pulmonary tissue. The attenuation of the recruitment of BM-derived c-kit+ cells by sildenafil may provide novel therapeutic insights into the control of pulmonary remodelling.


Sujet(s)
Cellules de la moelle osseuse/anatomopathologie , Poumon/anatomopathologie , Citrate de sildénafil/pharmacologie , Cellules souches/anatomopathologie , Animaux , Gazométrie sanguine , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/métabolisme , Hypoxie cellulaire/effets des médicaments et des substances chimiques , GMP cyclique/métabolisme , Inflammation/anatomopathologie , Mâle , Muscles/effets des médicaments et des substances chimiques , Muscles/anatomopathologie , Protéines proto-oncogènes c-kit/métabolisme , Rat Sprague-Dawley , Récepteurs CXCR4/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme
14.
Int J Cardiol ; 212: 324-35, 2016 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-27057951

RÉSUMÉ

BACKGROUND: Critical limb ischemia (CLI) is a major health problem with no adequate treatment. Since CLI is characterized by insufficient tissue vascularization, efforts have focused on the discovery of novel angiogenic factors. Cyclophilin A (CyPA) is an immunophilin that has been shown to promote angiogenesis in vitro and to enhance bone marrow (BM) cell mobilization in vivo. However, its potential as an angiogenic factor in CLI is still unknown. Thus, this study aimed to evaluate whether CyPA might induce neo-angiogenesis in ischemic tissues. METHODS AND RESULTS: Wild-type C57Bl/6j mice underwent acute hind-limb ischemia (HLI) and received a single intramuscular administration of recombinant CyPA or saline. Limb perfusion, capillary density and arteriole number in adductor muscles were significantly increased after CyPA treatment. Interestingly, BM-derived CD117(+) cell recruitment was significantly higher in ischemic adductor tissue of mice treated with CyPA versus saline. Therefore, the effect of CyPA on isolated BM-derived CD117(+) cells in vitro was evaluated. Low concentrations of CyPA stimulated CD117(+) cell proliferation while high concentrations promoted cell death. Moreover, CyPA enhanced CD117(+) cell adhesion and migration in a dose-dependent manner. Mechanistic studies revealed that CyPA up-regulated CXCR4 in CD117(+) cells and in adductor muscles after ischemia. Additionally, SDF-1/CXCR4 axis inhibition by the CXCR4 antagonist AMD3100 decreased CyPA-mediated CD117(+) cell recruitment in the ischemic limb. CONCLUSION: CyPA induces neo-angiogenesis by recruiting BM-derived CD117(+) cell into ischemic tissues, at least in part, through SDF-1/CXCR4 axis.


Sujet(s)
Cyclophiline A/administration et posologie , Membre pelvien/vascularisation , Ischémie/traitement médicamenteux , Néovascularisation physiologique/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-kit/métabolisme , Animaux , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Adhérence cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Chimiokine CXCL12/métabolisme , Cyclophiline A/pharmacologie , Modèles animaux de maladie humaine , Membre pelvien/effets des médicaments et des substances chimiques , Humains , Ischémie/métabolisme , Souris , Souris de lignée C57BL , Récepteurs CXCR4/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
15.
Stem Cells Int ; 2016: 4287158, 2016.
Article de Anglais | MEDLINE | ID: mdl-27110250

RÉSUMÉ

A mere 9 years have passed since the revolutionary report describing the derivation of induced pluripotent stem cells from human fibroblasts and the first in-patient translational use of cells obtained from these stem cells has already been achieved. From the perspectives of clinicians and researchers alike, the promise of induced pluripotent stem cells is alluring if somewhat beguiling. It is now evident that this technology is nascent and many areas for refinement have been identified and need to be considered before induced pluripotent stem cells can be routinely used to stratify, treat and cure patients, and to faithfully model diseases for drug screening purposes. This review specifically addresses the pioneering approaches to improve induced pluripotent stem cell based models of nonischaemic cardiomyopathy.

16.
Mitochondrion ; 27: 40-7, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26910457

RÉSUMÉ

The regulation and function of Mitochondrial DNA (mtDNA) cytosine methylation (5 mC) are largely unexplored. Mitochondria, Endothelial Cell (EC) senescence, and cardiovascular dysfunction are closely related. We extensively investigated the mtDNA Non-Coding Region (NCR) methylation pattern and its variations in EC replicative senescence. We observed previously undescribed 5 mC clusters and a biased distribution of 5 mC among DNA sites and throughout the NCR. The methylation pattern in senescent EC showed non-random variations, including the hypo-methylation of mtDNA replication regulatory sites. Additional experiments opened to a possible role for 5 mC in D-loop formation, rather than in mitochondrial gene expression.


Sujet(s)
Cytosine/analogues et dérivés , ADN intergénique/composition chimique , ADN mitochondrial/composition chimique , Cellules endothéliales/composition chimique , Mitochondries/composition chimique , 5-Méthyl-cytosine/analogues et dérivés , Cytosine/analyse , Humains , Analyse de séquence d'ADN/méthodes , Sulfites/métabolisme
17.
Cardiovasc Res ; 112(2): 555-567, 2016 Nov 01.
Article de Anglais | MEDLINE | ID: mdl-28513807

RÉSUMÉ

AIMS: Beyond the control of nuclear-cytoplasmic trafficking nucleoporins regulate gene expression and are involved in cardiac diseases. Notably, a number of cardiovascular disorders have been linked to alterations in epigenetic mechanisms. Here we aimed to determine the contribution of Nup153 to the epigenetic alterations occurring in cardiomyopathy of dystrophin-deficient mdx mice (C57BL/10ScSn-Dmd mdx /J). METHODS AND RESULTS: Nup153 was lysine-acetylated and its expression was significantly increased at protein level in mdx hearts compared with controls. Accordingly, lysine acetyl transferase (KAT) activity associated with Nup153 was higher in mdx hearts paralleling increased binding with the lysine acetylases P300/CBP-associated factor (PCAF) and p300. Interestingly, Nup153 silencing in mdx organotypic heart tissue slices caused a reduction in PCAF- and p300-specific activities. Remarkably, the level of nitric oxide (NO), which is reduced in mdx mice, was important for KAT-dependent regulation of Nup153. In fact, treatment of mdx heart tissue with an NO donor or the KAT inhibitor anacardic acid normalized Nup153 protein expression. Nup153 was recruited to chromatin and regulated the transcription of genes involved in cardiac remodelling, including the actin-binding protein nexilin. Accordingly, nexilin protein expression was abrogated by Nup153 silencing in mdx organotypic cultures. Electrophysiological and molecular experiments revealed that Nup153 overexpression in normal cardiomyocytes increases Ca v 1.2 calcium channel expression and function. Alterations in Nup153 protein expression and intracellular localization were also found in dystrophic cardiomyocytes derived from patient-specific induced pluripotent stem cells. Importantly, Nup153 up-regulation and increased acetylation were also found in the heart of Duchenne muscular dystrophy patients. CONCLUSIONS: Our data indicate that Nup153 is an epigenetic regulator which, upon altered NO signalling, mediates the activation of genes potentially associated with early dystrophic cardiac remodelling.


Sujet(s)
Chromatine/physiologie , Régulation de l'expression des gènes , Myopathie de Duchenne/métabolisme , Myocytes cardiaques/métabolisme , Complexe protéique du pore nucléaire/physiologie , Acétylation , Animaux , Canaux calciques/physiologie , Cellules cultivées , Humains , Mâle , Souris , Souris de lignée C57BL , Souris de lignée mdx , Monoxyde d'azote/métabolisme , Rats , Remodelage ventriculaire
19.
Cardiovasc Res ; 106(3): 353-64, 2015 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-25750190

RÉSUMÉ

Peptidyl-prolyl cis-trans-isomerases are a highly conserved family of immunophilins. The three peptidyl-prolyl cis-trans-isomerase subfamilies are cyclophilins, FK-506-binding proteins, and parvulins. Peptidyl-prolyl cis-trans-isomerases are expressed in multiple human tissues and regulate different cellular functions, e.g. calcium handling, protein folding, and gene expression. Moreover, these subfamilies have been shown to be consistently involved in several cardiac and vascular diseases including heart failure, arrhythmias, vascular stenosis, endothelial dysfunction, atherosclerosis, and hypertension. This review provides a concise description of the peptidyl-prolyl cis-trans-isomerases and presents an incisive selection of studies focused on their relationship with cardiovascular diseases.


Sujet(s)
Maladies cardiovasculaires/enzymologie , Système cardiovasculaire/enzymologie , Peptidylpropyl isomerase/métabolisme , Transduction du signal , Animaux , Agents cardiovasculaires/usage thérapeutique , Maladies cardiovasculaires/traitement médicamenteux , Maladies cardiovasculaires/physiopathologie , Système cardiovasculaire/effets des médicaments et des substances chimiques , Système cardiovasculaire/physiopathologie , Cyclophilines/métabolisme , Antienzymes/usage thérapeutique , Humains , Thérapie moléculaire ciblée , NIMA-interacting peptidylprolyl isomerase , Peptidylpropyl isomerase/antagonistes et inhibiteurs , Transduction du signal/effets des médicaments et des substances chimiques , Protéines de liaison au tacrolimus/métabolisme
20.
J Mol Cell Cardiol ; 81: 62-70, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25640160

RÉSUMÉ

Age-associated cardiovascular diseases are at least partially ascribable to vascular cell senescence. Replicative senescence (RS) and stress-induced premature senescence (SIPS) are provoked respectively by endogenous (telomere erosion) and exogenous (H2O2, UV) stimuli resulting in cell cycle arrest in G1 and G2 phases. In both scenarios, mitochondria-derived ROS are important players in senescence initiation. We aimed to define whether a mtDNA-transcribed long-non-coding-RNA (lncRNA), ASncmtRNA-2, has a role in vascular aging and senescence. Aortas of old mice, characterized by increased senescence, showed an increment in ASncmtRNA-2 expression. In vitro analysis of Endothelial Cells (EC) and Vascular Smooth Muscle Cells (VSMC) established that ASncmtRNA-2 is induced in EC, but not in VSMC, during RS. Surprisingly, ASncmtRNA-2 is not upregulated in two different EC SIPS scenarios, treated with H2O2 and UV. The p16 gene displayed similar ASncmtRNA-2 expression patterns, suggesting a possible co-regulation of the two genes. Interestingly, the expression of two miRNAs, hsa-miR-4485 and hsa-miR-1973, with perfect homology to the double strand region of ASncmtRNA-2 and originating at least in part from a mitochondrial transcript, was induced in RS, opening to the possibility that this lncRNA functions as a non-canonical precursor of these miRNAs. Cell cycle analysis of EC transiently over-expressing ASncmtRNA-2 revealed an accumulation of EC in the G2/M phase, but not in the G1 phase. We propose that ASncmtRNA-2 in EC might be involved in the RS establishment by participating in the cell cycle arrest in G2/M phase, possibly through the production of hsa-miR-4485 and hsa-miR-1973. This article is part of a Special Issue entitled: Mitochondria.


Sujet(s)
Vieillissement/métabolisme , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Mitochondries/métabolisme , Myocytes du muscle lisse/métabolisme , ARN long non codant/génétique , ARN/génétique , Vieillissement/génétique , Animaux , Aorte/cytologie , Aorte/métabolisme , Séquence nucléotidique , Vieillissement de la cellule , Inhibiteur p16 de kinase cycline-dépendante/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Points de contrôle de la phase G2 du cycle cellulaire/effets des médicaments et des substances chimiques , Points de contrôle de la phase G2 du cycle cellulaire/effets des radiations , Régulation de l'expression des gènes , Cellules endothéliales de la veine ombilicale humaine/cytologie , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des radiations , Humains , Peroxyde d'hydrogène/pharmacologie , Mâle , Souris , Souris de lignée C57BL , microARN/génétique , microARN/métabolisme , Mitochondries/génétique , Données de séquences moléculaires , Myocytes du muscle lisse/cytologie , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/effets des radiations , ARN/métabolisme , ARN long non codant/métabolisme , ARN mitochondrial , Transduction du signal , Rayons ultraviolets
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...