Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 31
Filtrer
1.
J Pharmacol Sci ; 148(2): 214-220, 2022 Feb.
Article de Anglais | MEDLINE | ID: mdl-35063136

RÉSUMÉ

Pulmonary hypertension (PH) is a severe and progressive disease that causes elevated right ventricular systolic pressure, right ventricular hypertrophy and ultimately right heart failure. However, the underlying pathophysiologic mechanisms are poorly understood. We previously showed that 3,4-l-dihydroxylphenyalanine (DOPA) sensitizes vasomotor response to sympathetic tone via coupling between the adrenergic receptor alpha1 (ADRA1) and a G protein-coupled receptor 143 (GPR143), a DOPA receptor. We investigated whether DOPA similarly enhances ADRA1-mediated contraction in pulmonary arteries isolated from rats, and whether GPR143 is involved in the PH pathogenesis. Pretreating the isolated pulmonary arteries with DOPA 1 µM enhanced vasoconstriction in response to phenylephrine, an ADRA1 agonist, but not to U-46619, a thromboxane A2 agonist or endothelin-1. We generated Gpr143 gene-deficient (Gpr143-/y) rats, and confirmed that DOPA did not augment phenylephrine-induced contractile response in Gpr143-/y rat pulmonary arteries. We utilized a rat model of monocrotaline (MCT)-induced PH. In the MCT model, the right ventricular systolic pressure was attenuated in the Gpr143-/y rats than in WT rats. Phenylephrine-induced cell migration and proliferation were also suppressed in Gpr143-/y pulmonary artery smooth muscle cells than in WT cells. Our result suggests that GPR143 is involved in the PH pathogenesis in the rat models of PH.


Sujet(s)
Hypertension pulmonaire/étiologie , Hypertension pulmonaire/génétique , Monocrotaline/effets indésirables , Récepteurs couplés aux protéines G/physiologie , Récepteurs aux neuromédiateurs/génétique , Systole , Fonction ventriculaire droite/génétique , Acide 15-hydroxy-11alpha,9alpha-(époxyméthano)prosta-5,13-diénoïque/pharmacologie , Animaux , Modèles animaux de maladie humaine , Défaillance cardiaque/étiologie , Hypertrophie ventriculaire droite/étiologie , Techniques in vitro , Mâle , Artère pulmonaire/physiologie , Rat Sprague-Dawley , Récepteurs alpha-1 adrénergiques/physiologie , Vasoconstriction/effets des médicaments et des substances chimiques , Vasoconstriction/génétique , Dysfonction ventriculaire droite/étiologie
2.
J Am Heart Assoc ; 10(16): e020854, 2021 08 17.
Article de Anglais | MEDLINE | ID: mdl-34387124

RÉSUMÉ

Background Current right ventricular (RV) volume overload (VO) is established in adult mice. There are no neonatal mouse VO models and how VO affects postnatal RV development is largely unknown. Methods and Results Neonatal VO was induced by the fistula between abdominal aorta and inferior vena cava on postnatal day 7 and confirmed by abdominal ultrasound, echocardiography, and hematoxylin and eosin staining. The RNA-sequencing results showed that the top 5 most enriched gene ontology terms in normal RV development were energy derivation by oxidation of organic compounds, generation of precursor metabolites and energy, cellular respiration, striated muscle tissue development, and muscle organ development. Under the influence of VO, the top 5 most enriched gene ontology terms were angiogenesis, regulation of cytoskeleton organization, regulation of vasculature development, regulation of mitotic cell cycle, and regulation of the actin filament-based process. The top 3 enriched signaling pathways for the normal RV development were PPAR signaling pathway, citrate cycle (Tricarboxylic acid cycle), and fatty acid degradation. VO changed the signaling pathways to focal adhesion, the PI3K-Akt signaling pathway, and pathways in cancer. The RNA sequencing results were confirmed by the examination of the markers of metabolic and cardiac muscle maturation and the markers of cell cycle and angiogenesis. Conclusions A neonatal mouse VO model was successfully established, and the main processes of postnatal RV development were metabolic and cardiac muscle maturation, and VO changed that to angiogenesis and cell cycle regulation.


Sujet(s)
Transcriptome , Dysfonction ventriculaire droite/génétique , Fonction ventriculaire droite/génétique , Animaux , Animaux nouveau-nés , Aorte abdominale/physiopathologie , Aorte abdominale/chirurgie , Anastomose chirurgicale artérioveineuse , Modèles animaux de maladie humaine , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes au cours du développement , Mâle , Souris de lignée C57BL , RNA-Seq , Facteurs temps , Veine cave inférieure/physiopathologie , Veine cave inférieure/chirurgie , Dysfonction ventriculaire droite/étiologie , Dysfonction ventriculaire droite/physiopathologie
3.
Am J Respir Cell Mol Biol ; 65(3): 272-287, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-33938785

RÉSUMÉ

Right ventricular (RV) function is the predominant determinant of survival in patients with pulmonary arterial hypertension (PAH). In preclinical models, pharmacological activation of BMP (bone morphogenetic protein) signaling with FK506 (tacrolimus) improved RV function by decreasing RV afterload. FK506 therapy further stabilized three patients with end-stage PAH. Whether FK506 has direct effects on the pressure-overloaded right ventricle is yet unknown. We hypothesized that increasing cardiac BMP signaling with FK506 improves RV structure and function in a model of fixed RV afterload after pulmonary artery banding (PAB). Direct cardiac effects of FK506 on the microvasculature and RV fibrosis were studied after surgical PAB in wild-type and heterozygous Bmpr2 mutant mice. RV function and strain were assessed longitudinally via cardiac magnetic resonance imaging during continuous FK506 infusion. Genetic lineage tracing of endothelial cells (ECs) was performed to assess the contribution of ECs to fibrosis. Molecular mechanistic studies were performed in human cardiac fibroblasts and ECs. In mice, low BMP signaling in the right ventricle exaggerated PAB-induced RV fibrosis. FK506 therapy restored cardiac BMP signaling, reduced RV fibrosis in a BMP-dependent manner independent from its immunosuppressive effect, preserved RV capillarization, and improved RV function and strain over the time course of disease. Endothelial mesenchymal transition was a rare event and did not significantly contribute to cardiac fibrosis after PAB. Mechanistically, FK506 required ALK1 in human cardiac fibroblasts as a BMPR2 co-receptor to reduce TGFß1-induced proliferation and collagen production. Our study demonstrates that increasing cardiac BMP signaling with FK506 improves RV structure and function independent from its previously described beneficial effects on pulmonary vascular remodeling.


Sujet(s)
Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Protéines morphogénétiques osseuses/métabolisme , Hypertension artérielle pulmonaire/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Tacrolimus/pharmacologie , Fonction ventriculaire droite/effets des médicaments et des substances chimiques , Animaux , Récepteurs de la protéine morphogénique osseuse de type II/génétique , Protéines morphogénétiques osseuses/génétique , Fibroblastes/métabolisme , Fibrose , Humains , Mâle , Souris , Souches mutantes de souris , Myocarde/métabolisme , Hypertension artérielle pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire/génétique , Transduction du signal/génétique , Fonction ventriculaire droite/génétique
4.
Sci Rep ; 11(1): 3587, 2021 02 11.
Article de Anglais | MEDLINE | ID: mdl-33574400

RÉSUMÉ

Right ventricular (RV) remodeling and longitudinal fiber reorientation in the setting of pulmonary hypertension (PH) affects ventricular structure and function, eventually leading to RV failure. Characterizing the kinematics of myocardial fibers helps better understanding the underlying mechanisms of fiber realignment in PH. In the current work, high-frequency ultrasound imaging and structurally-informed finite element (FE) models were employed for an exploratory evaluation of the stretch-induced kinematics of RV fibers. Image-based experimental evaluation of fiber kinematics in porcine myocardium revealed the capability of affine assumptions to effectively approximate myofiber realignment in the RV free wall. The developed imaging framework provides a noninvasive modality to quantify transmural RV myofiber kinematics in large animal models. FE modeling results demonstrated that chronic pressure overload, but not solely an acute rise in pressures, results in kinematic shift of RV fibers towards the longitudinal direction. Additionally, FE simulations suggest a potential protective role for concentric hypertrophy (increased wall thickness) against fiber reorientation, while eccentric hypertrophy (RV dilation) resulted in longitudinal fiber realignment. Our study improves the current understanding of the role of different remodeling events involved in transmural myofiber reorientation in PH. Future experimentations are warranted to test the model-generated hypotheses.


Sujet(s)
Ventricules cardiaques/imagerie diagnostique , Hypertension pulmonaire/imagerie diagnostique , Dysfonction ventriculaire droite/imagerie diagnostique , Remodelage ventriculaire/physiologie , Animaux , Phénomènes biomécaniques , Modèles animaux de maladie humaine , Ventricules cardiaques/physiopathologie , Humains , Hypertension pulmonaire/physiopathologie , Hypertrophie ventriculaire droite/imagerie diagnostique , Hypertrophie ventriculaire droite/génétique , Hypertrophie ventriculaire droite/physiopathologie , Myocytes cardiaques/anatomopathologie , Suidae , Dysfonction ventriculaire droite/génétique , Dysfonction ventriculaire droite/physiopathologie , Fonction ventriculaire droite/génétique , Fonction ventriculaire droite/physiologie , Pression ventriculaire/physiologie , Remodelage ventriculaire/génétique
5.
Sci Rep ; 11(1): 2012, 2021 01 21.
Article de Anglais | MEDLINE | ID: mdl-33479312

RÉSUMÉ

Despite the advancement of targeted therapy for pulmonary arterial hypertension (PAH), poor prognosis remains a reality. Mesenchymal stem cells (MSCs) are one of the most clinically feasible alternative treatment options. We compared the treatment effects of adipose tissue (AD)-, bone marrow (BD)-, and umbilical cord blood (UCB)-derived MSCs in the rat monocrotaline-induced pulmonary hypertension (PH) model. The greatest improvement in the right ventricular function was observed in the UCB-MSCs treated group. The UCB-MSCs treated group also exhibited the greatest improvement in terms of the largest decrease in the medial wall thickness, perivascular fibrosis, and vascular cell proliferation, as well as the lowest levels of recruitment of innate and adaptive immune cells and associated inflammatory cytokines. Gene expression profiling of lung tissue confirmed that the UCB-MSCs treated group had the most notably attenuated immune and inflammatory profiles. Network analysis further revealed that the UCB-MSCs group had the greatest therapeutic effect in terms of the normalization of all three classical PAH pathways. The intravenous injection of the UCB-MSCs, compared with those of other MSCs, showed superior therapeutic effects in the PH model for the (1) right ventricular function, (2) vascular remodeling, (3) immune/inflammatory profiles, and (4) classical PAH pathways.


Sujet(s)
Thérapie cellulaire et tissulaire , Transplantation de cellules souches mésenchymateuses , Hypertension artérielle pulmonaire/thérapie , Remodelage vasculaire/génétique , Animaux , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/métabolisme , Prolifération cellulaire/génétique , Transplantation de cellules souches de sang du cordon , Modèles animaux de maladie humaine , Régulation de l'expression des gènes/génétique , Humains , Cellules souches mésenchymateuses/cytologie , Cellules souches mésenchymateuses/métabolisme , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/anatomopathologie , Artère pulmonaire/croissance et développement , Artère pulmonaire/anatomopathologie , Rats , Fonction ventriculaire droite/génétique
6.
Int J Mol Sci ; 21(23)2020 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-33255338

RÉSUMÉ

Pulmonary artery hypertension (PAH) is a rare chronic disease with high impact on patients' quality of life and currently no available cure. PAH is characterized by constant remodeling of the pulmonary artery by increased proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs), fibroblasts (FBs) and endothelial cells (ECs). This remodeling eventually leads to increased pressure in the right ventricle (RV) and subsequent right ventricle hypertrophy (RVH) which, when left untreated, progresses into right ventricle failure (RVF). PAH can not only originate from heritable mutations, but also develop as a consequence of congenital heart disease, exposure to drugs or toxins, HIV, connective tissue disease or be idiopathic. While much attention was drawn into investigating and developing therapies related to the most well understood signaling pathways in PAH, in the last decade, a shift towards understanding the epigenetic mechanisms driving the disease occurred. In this review, we reflect on the different epigenetic regulatory factors that are associated with the pathology of RV remodeling, and on their relevance towards a better understanding of the disease and subsequently, the development of new and more efficient therapeutic strategies.


Sujet(s)
Épigenèse génétique/génétique , Hypertension artérielle pulmonaire/génétique , Dysfonction ventriculaire droite/génétique , Remodelage ventriculaire/génétique , Animaux , Modèles animaux de maladie humaine , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Humains , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Hypertension artérielle pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Transduction du signal , Dysfonction ventriculaire droite/anatomopathologie , Fonction ventriculaire droite/génétique
7.
J Am Heart Assoc ; 9(16): e015342, 2020 08 18.
Article de Anglais | MEDLINE | ID: mdl-32805183

RÉSUMÉ

Background Patients with repair of tetralogy of Fallot (rToF) who are approaching adulthood often exhibit pulmonary valve regurgitation, leading to right ventricle (RV) dilatation and dysfunction. The regurgitation can be corrected by pulmonary valve replacement (PVR), but the optimal surgical timing remains under debate, mainly because of the poorly understood nature of RV remodeling in patients with rToF. The goal of this study was to probe for pathologic molecular, cellular, and tissue changes in the myocardium of patients with rToF at the time of PVR. Methods and Results We measured contractile function of permeabilized myocytes, collagen content of tissue samples, and the expression of mRNA and selected proteins in RV tissue samples from patients with rToF undergoing PVR for severe pulmonary valve regurgitation. The data were compared with nondiseased RV tissue from unused donor hearts. Contractile performance and passive stiffness of the myofilaments in permeabilized myocytes were similar in rToF-PVR and RV donor samples, as was collagen content and cross-linking. The patients with rToF undergoing PVR had enhanced mRNA expression of genes associated with connective tissue diseases and tissue remodeling, including the small leucine-rich proteoglycans ASPN (asporin), LUM (lumican), and OGN (osteoglycin), although their protein levels were not significantly increased. Conclusions RV myofilaments from patients with rToF undergoing PVR showed no functional impairment, but the changes in extracellular matrix gene expression may indicate the early stages of remodeling. Our study found no evidence of major damage at the cellular and tissue levels in the RV of patients with rToF who underwent PVR according to current clinical criteria.


Sujet(s)
Matrice extracellulaire/génétique , Expression des gènes , Myocytes cardiaques/physiologie , Myofibrilles/physiologie , Tétralogie de Fallot/génétique , Fonction ventriculaire droite/génétique , Adolescent , Adulte , Enfant , Collagène/analyse , Régulation négative , Protéines de la matrice extracellulaire/isolement et purification , Femelle , Analyse de profil d'expression de gènes/méthodes , Implantation de valve prothétique cardiaque , Humains , Mâle , Adulte d'âge moyen , Contraction musculaire/physiologie , Réaction de polymérisation en chaîne , Valve du tronc pulmonaire/chirurgie , Insuffisance pulmonaire/chirurgie , ARN messager/métabolisme , Petits protéoglycanes riches en leucine/métabolisme , Tétralogie de Fallot/chirurgie , Régulation positive , Jeune adulte
8.
Am J Respir Cell Mol Biol ; 63(5): 652-664, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32692928

RÉSUMÉ

Pulmonary hypertension (PH) and right ventricular (RV) hypertrophy frequently develop in patients with hypoxic lung disease. Chronic alveolar hypoxia (CH) promotes sustained pulmonary vasoconstriction and pulmonary artery (PA) remodeling by acting on lung cells, resulting in the development of PH. RV hypertrophy develops in response to PH, but coronary arterial hypoxemia in CH may influence that response by activating HIF-1α (hypoxia-inducible factor 1α) and/or HIF-2α in cardiomyocytes. Indeed, other studies show that the attenuation of PH in CH fails to prevent RV remodeling, suggesting that PH-independent factors regulate RV hypertrophy. Therefore, we examined the role of HIFs in RV remodeling in CH-induced PH. We deleted HIF-1α and/or HIF-2α in hearts of adult mice that were then housed under normoxia or CH (10% O2) for 4 weeks. RNA-sequencing analysis of the RV revealed that HIF-1α and HIF-2α regulate the transcription of largely distinct gene sets during CH. RV systolic pressure increased, and RV hypertrophy developed in CH. The deletion of HIF-1α in smooth muscle attenuated the CH-induced increases in RV systolic pressure but did not decrease hypertrophy. The deletion of HIF-1α in cardiomyocytes amplified RV remodeling; this was abrogated by the simultaneous loss of HIF-2α. CH decreased stroke volume and cardiac output in wild-type but not in HIF-1α-deficient hearts, suggesting that CH may cause cardiac dysfunction via HIF-dependent signaling. Collectively, these data reveal that HIF-1 and HIF-2 act together in RV cardiomyocytes to orchestrate RV remodeling in CH, with HIF-1 playing a protective role rather than driving hypertrophy.


Sujet(s)
Hypertension pulmonaire/étiologie , Hypertension pulmonaire/physiopathologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Hypoxie/complications , Fonction ventriculaire droite/physiologie , Remodelage ventriculaire/physiologie , Animaux , Maladie chronique , Délétion de gène , Régulation de l'expression des gènes , Gene Ontology , Hypertension pulmonaire/génétique , Integrases/métabolisme , Souris , Myocytes cardiaques/métabolisme , Artère pulmonaire/anatomopathologie , Artère pulmonaire/physiopathologie , Transcription génétique , Fonction ventriculaire droite/génétique , Remodelage ventriculaire/génétique
9.
J Am Heart Assoc ; 9(11): e015574, 2020 06 02.
Article de Anglais | MEDLINE | ID: mdl-32475201

RÉSUMÉ

Background Current mammalian models for heart regeneration research are limited to neonatal apex amputation and myocardial infarction, both of which are controversial. RNAseq has demonstrated a very limited set of differentially expressed genes between sham and operated hearts in myocardial infarction models. Here, we investigated in rats whether pressure overload in the right ventricle, a common phenomenon in children with congenital heart disease, could be used as a better animal model for heart regeneration studies when considering cardiomyocyte proliferation as the most important index. Methods and Results In the rat model, pressure overload was induced by pulmonary artery banding on postnatal day 1 and confirmed by echocardiography and hemodynamic measurements at postnatal day 7. RNA sequencing analyses of purified right ventricular cardiomyocytes at postnatal day 7 from pulmonary artery banding and sham-operated rats revealed that there were 5469 differentially expressed genes between these 2 groups. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analysis showed that these genes mainly mediated mitosis and cell division. Cell proliferation assays indicated a continuous overproliferation of cardiomyocytes in the right ventricle after pulmonary artery banding, in particular for the first 3 postnatal days. We also validated the model using samples from overloaded right ventricles of human patients. There was an approximately 2-fold increase of Ki67/pHH3/aurora B-positive cardiomyocytes in human-overloaded right ventricles compared with nonoverloaded right ventricles. Other features of this animal model included cardiomyocyte hypotrophy with no fibrosis. Conclusions Pressure overload profoundly promotes cardiomyocyte proliferation in the neonatal stage in both rats and human beings. This activates a regeneration-specific gene program and may offer an alternative animal model for heart regeneration research.


Sujet(s)
Prolifération cellulaire , Hypertrophie ventriculaire droite/anatomopathologie , Myocytes cardiaques/anatomopathologie , Régénération , Fonction ventriculaire droite , Pression ventriculaire , Remodelage ventriculaire , Animaux , Animaux nouveau-nés , Modèles animaux de maladie humaine , Femelle , Fibrose , Analyse de profil d'expression de gènes , Réseaux de régulation génique , Défaillance cardiaque/étiologie , Défaillance cardiaque/génétique , Défaillance cardiaque/anatomopathologie , Défaillance cardiaque/physiopathologie , Humains , Hypertrophie ventriculaire droite/étiologie , Hypertrophie ventriculaire droite/génétique , Hypertrophie ventriculaire droite/physiopathologie , Mâle , Myocytes cardiaques/métabolisme , Artère pulmonaire/physiopathologie , Artère pulmonaire/chirurgie , RNA-Seq , Rat Sprague-Dawley , Régénération/génétique , Tétralogie de Fallot/génétique , Tétralogie de Fallot/anatomopathologie , Tétralogie de Fallot/physiopathologie , Facteurs temps , Transcriptome , Fonction ventriculaire droite/génétique , Pression ventriculaire/génétique , Remodelage ventriculaire/génétique
10.
BMC Cardiovasc Disord ; 20(1): 74, 2020 02 11.
Article de Anglais | MEDLINE | ID: mdl-32046637

RÉSUMÉ

BACKGROUND: Arrhythmogenic cardiomyopathy (AC) is one of the leading causes for sudden cardiac death (SCD). Recent studies have identified mutations in cardiac desmosomes as key players in the pathogenesis of AC. However, the specific etiology in individual families remains largely unknown. METHODS: A 4-generation family presenting with syncope, lethal ventricular arrhythmia and SCD was recruited. Targeted next generation sequencing (NGS) was performed and validated by Sanger sequencing. Plasmids containing the mutation and wild type (WT) were constructed. Real-time PCR, western-blot and immunofluorescence were performed to detect the functional change due to the mutation. RESULTS: The proband, a 56-year-old female, presented with recurrent palpitations and syncope. An ICD was implanted due to her family history of SCD/ aborted SCD. NGS revealed a novel heterozygous frame-shift variant (c.832delG) in Desmoplakin (DSP) among 5 family members. The variant led to frame-shift and premature termination, producing a truncated protein. Cardiac magnetic resonance (CMR) of the family members carrying the same variant shown myocardium thinning and fatty infiltration in the right ventricular, positive bi-ventricular late gadolinium enhancement and severe RV dysfunction, fulfilling the diagnostic criteria of AC. HEK293T cells transfected with mutant plasmids expressed truncated DSP mRNA and protein, upregulation of nuclear junction plakoglobin (JUP) and downregulation of ß-catenin, when compared with WT. CONCLUSION: We infer that the novel c.832delG variant in DSP was associated with AC in this family, likely through Wnt/ß-catenin signaling pathway.


Sujet(s)
Troubles du rythme cardiaque/génétique , Cardiomyopathies/génétique , Analyse de mutations d'ADN , Desmoplakines/génétique , Mutation avec décalage du cadre de lecture , Séquençage nucléotidique à haut débit , Adolescent , Adulte , Sujet âgé de 80 ans ou plus , Troubles du rythme cardiaque/imagerie diagnostique , Troubles du rythme cardiaque/métabolisme , Troubles du rythme cardiaque/physiopathologie , Cardiomyopathies/imagerie diagnostique , Cardiomyopathies/métabolisme , Cardiomyopathies/physiopathologie , Mort subite cardiaque/étiologie , Desmoplakines/métabolisme , Femelle , Prédisposition génétique à une maladie , Cellules HEK293 , Humains , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Phénotype , Valeur prédictive des tests , Fonction ventriculaire droite/génétique , Jeune adulte , bêta-Caténine/métabolisme , gamma-Caténine/métabolisme
12.
JACC Cardiovasc Imaging ; 12(3): 446-455, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-29550307

RÉSUMÉ

OBJECTIVES: The aim of this study was to investigate the prognostic value of echocardiographic deformation imaging in arrhythmogenic right ventricular cardiomyopathy (ARVC) to optimize family screening protocols. BACKGROUND: ARVC is characterized by variable disease expressivity among family members, which complicates family screening protocols. Previous reports have shown that echocardiographic deformation imaging detects abnormal right ventricular (RV) deformation in the absence of established disease expression in ARVC. METHODS: First-degree relatives of patients with ARVC were evaluated according to 2010 task force criteria, including RV deformation imaging (n = 128). Relatives fulfilling structural task force criteria were excluded for further analysis. At baseline, deformation patterns of the subtricuspid region were scored as type I (normal deformation), type II (delayed onset, decreased systolic peak, and post-systolic shortening), or type III (systolic stretching and large post-systolic shortening). The final study population comprised relatives who underwent a second evaluation during follow-up. Disease progression was defined as the development of a new 2010 task force criterion during follow-up that was absent at baseline. RESULTS: Sixty-five relatives underwent a second evaluation after a mean follow-up period of 3.7 ± 2.1 years. At baseline, 28 relatives (43%) had normal deformation (type I), and 37 relatives (57%) had abnormal deformation (type II or III) in the subtricuspid region. Disease progression occurred in 4% of the relatives with normal deformation at baseline and in 43% of the relatives with abnormal deformation at baseline (p < 0.001). Positive and negative predictive values of abnormal deformation were, respectively, 43% (95% confidence interval: 27% to 61%) and 96% (95% confidence interval: 82% to 100%). CONCLUSIONS: Normal RV deformation in the subtricuspid region is associated with absence of disease progression during nearly 4-year follow-up in relatives of patients with ARVC. Abnormal RV deformation seems to precede the established signs of ARVC. RV deformation imaging may potentially play an important role in ARVC family screening protocols.


Sujet(s)
Dysplasie ventriculaire droite arythmogène/imagerie diagnostique , Échocardiographie-doppler , Imagerie par résonance magnétique , Contraction myocardique , Fonction ventriculaire droite , Adolescent , Adulte , Dysplasie ventriculaire droite arythmogène/génétique , Dysplasie ventriculaire droite arythmogène/physiopathologie , Évolution de la maladie , Diagnostic précoce , Électrocardiographie , Femelle , Prédisposition génétique à une maladie , Hérédité , Humains , Mâle , Adulte d'âge moyen , Contraction myocardique/génétique , Pedigree , Phénotype , Valeur prédictive des tests , Pronostic , Enregistrements , Fonction ventriculaire droite/génétique , Jeune adulte
13.
Sarcoidosis Vasc Diffuse Lung Dis ; 36(4): 285-293, 2019.
Article de Anglais | MEDLINE | ID: mdl-32476964

RÉSUMÉ

BACKGROUND: Impaired systolic function is common in sarcoidosis however the frequency of diastolic dysfunction (DD) and it's possible genetic basis has not been fully elucidated yet. The aim of this study is to evaluate the frequency of left ventricular DD(LVDD) and right ventricular DD(RVDD) and it's possible relationship between Human Leukocyte Antigen(HLA)-DRB1* alleles in patients with sarcoidosis. METHODS: Seventy seven patients (51 females, mean age 41.1±8.2yrs) without known sarcoid related or any other structured heart disease and 77 healthy controls with a similar age and gender (38.7±7.8yrs,51 females) were included in the case control study. DD was diagnosed with echocardiography. RVDD was defined as early(E)/late(A) ratio<1 or >2 on tricuspit valve. LVDD was defined as E/A ratio<1 or >2 on mitral valve, with isovolumetric relaxation time(IVRT)>90 miliseconds(msn) or deceleration rate of early diastolic flow(Edec)>220msn respectively. All patients were HLAtyped with the Sequence Specific Oligonucleotide Probe(SSOP) method. RESULTS: The frequencies of LVDDs and RVDDs were significantly higher in sarcoidosis patients than the controls (26.0% vs. 2.6% for LVDD; and 42.9% vs. 18.2% for RVDD)(p<0.05). No significant difference was found in patients according to the presence of RVDD and LVDD in terms of age, gender or respiratory function test parameters. Although the frequency of HLA DRB1* alleles were comparable among patients with RVDD, HLA DRB1*14 alleles were more frequent in patients with LVDD. CONCLUSIONS: Biventricular DD is common in patients with sarcoidosis without manifest cardiac involvement. HLA DRB1*14 allele seems to be related with LVDD in this study population.


Sujet(s)
Chaines HLA-DRB1/génétique , Sarcoïdose pulmonaire/complications , Dysfonction ventriculaire gauche/étiologie , Dysfonction ventriculaire droite/étiologie , Fonction ventriculaire gauche/génétique , Fonction ventriculaire droite/génétique , Adulte , Études cas-témoins , Diastole , Femelle , Fréquence d'allèle , Prédisposition génétique à une maladie , Humains , Poumon/physiopathologie , Mâle , Adulte d'âge moyen , Phénotype , Facteurs de risque , Sarcoïdose pulmonaire/diagnostic , Sarcoïdose pulmonaire/génétique , Sarcoïdose pulmonaire/physiopathologie , Dysfonction ventriculaire gauche/imagerie diagnostique , Dysfonction ventriculaire gauche/génétique , Dysfonction ventriculaire gauche/physiopathologie , Dysfonction ventriculaire droite/imagerie diagnostique , Dysfonction ventriculaire droite/génétique , Dysfonction ventriculaire droite/physiopathologie
14.
Circulation ; 139(7): 932-948, 2019 02 12.
Article de Anglais | MEDLINE | ID: mdl-30586714

RÉSUMÉ

BACKGROUND: Monoallelic mutations in the gene encoding bone morphogenetic protein receptor 2 ( Bmpr2) are the main genetic risk factor for heritable pulmonary arterial hypertension (PAH) with incomplete penetrance. Several Bmpr2 transgenic mice have been reported to develop mild spontaneous PAH. In this study, we examined whether rats with the Bmpr2 mutation were susceptible to developing more severe PAH. METHODS: The zinc finger nuclease method was used to establish rat lines with mutations in the Bmpr2 gene. These rats were then characterized at the hemodynamic, histological, electrophysiological, and molecular levels. RESULTS: Rats with a monoallelic deletion of 71 bp in exon 1 (Δ 71 rats) showed decreased BMPRII expression and phosphorylated SMAD1/5/9 levels. Δ 71 Rats develop age-dependent spontaneous PAH with a low penetrance (16%-27%), similar to that in humans. Δ 71 Rats were more susceptible to hypoxia-induced pulmonary hypertension than wild-type rats. Δ 71 Rats exhibited progressive pulmonary vascular remodeling associated with a proproliferative phenotype and showed lower pulmonary microvascular density than wild-type rats. Organ bath studies revealed severe alteration of pulmonary artery contraction and relaxation associated with potassium channel subfamily K member 3 (KCNK3) dysfunction. High levels of perivascular fibrillar collagen and pulmonary interleukin-6 overexpression discriminated rats that developed spontaneous PAH and rats that did not develop spontaneous PAH. Finally, detailed assessments of cardiomyocytes demonstrated alterations in morphology, calcium (Ca2+), and cell contractility specific to the right ventricle; these changes could explain the lower cardiac output of Δ 71 rats. Indeed, adult right ventricular cardiomyocytes from Δ 71 rats exhibited a smaller diameter, decreased sensitivity of sarcomeres to Ca2+, decreased [Ca2+] transient amplitude, reduced sarcoplasmic reticulum Ca2+ content, and short action potential duration compared with right ventricular cardiomyocytes from wild-type rats. CONCLUSIONS: We characterized the first Bmpr2 mutant rats and showed some of the critical cellular and molecular dysfunctions described in human PAH. We also identified the heart as an unexpected but potential target organ of Bmpr2 mutations. Thus, this new genetic rat model represents a promising tool to study the pathogenesis of PAH.


Sujet(s)
Pression artérielle/génétique , Récepteurs de la protéine morphogénique osseuse de type II/génétique , Hypertension pulmonaire/génétique , Hypertension pulmonaire/physiopathologie , Mutation , Contraction myocardique/génétique , Artère pulmonaire/physiopathologie , Fonction ventriculaire droite/génétique , Potentiels d'action , Animaux , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Signalisation calcique , Modèles animaux de maladie humaine , Prédisposition génétique à une maladie , Hypertension pulmonaire/métabolisme , Hypoxie/complications , Myocytes cardiaques/métabolisme , Protéines de tissu nerveux/métabolisme , Phénotype , Phosphorylation , Canaux potassiques à pores à domaines en tandem/métabolisme , Artère pulmonaire/métabolisme , Souches mutantes de rat , Protéines Smad/métabolisme
15.
Am J Physiol Heart Circ Physiol ; 315(4): H847-H854, 2018 10 01.
Article de Anglais | MEDLINE | ID: mdl-29906222

RÉSUMÉ

The right ventricular (RV) response to pulmonary arterial hypertension (PAH) is heterogeneous. Most patients have maladaptive changes with RV dilation and RV failure, whereas some, especially patients with PAH secondary to congenital heart disease, have an adaptive response with hypertrophy and preserved systolic function. Mechanisms for RV adaptation to PAH are unknown, despite RV function being a primary determinant of mortality. In our congenital heart disease ovine model with fetally implanted aortopulmonary shunt (shunt lambs), we previously demonstrated an adaptive physiological RV response to increased afterload with hypertrophy. In the present study, we examined small noncoding microRNA (miRNA) expression in shunt RV and characterized downstream effects of a key miRNA. RV tissue was harvested from 4-wk-old shunt and control lambs ( n = 5), and miRNA, mRNA, and protein were quantitated. We found differential expression of 40 cardiovascular-specific miRNAs in shunt RV. Interestingly, this miRNA signature is distinct from models of RV failure, suggesting that miRNAs might contribute to adaptive RV hypertrophy. Among RV miRNAs, miR-199b was decreased in the RV with eventual downregulation of nuclear factor of activated T cells/calcineurin signaling. Furthermore, antifibrotic miR-29a was increased in the shunt RV with a reduction of the miR-29 targets collagen type A1 and type 3A1 and decreased fibrosis. Thus, we conclude that the miRNA signature specific to shunt lambs is distinct from RV failure and drives gene expression required for adaptive RV hypertrophy. We propose that the adaptive RV miRNA signature may serve as a prognostic and therapeutic tool in patients with PAH to attenuate or prevent progression of RV failure and premature death. NEW & NOTEWORTHY This study describes a novel microRNA signature of adaptive right ventricular hypertrophy, with particular attention to miR-199b and miR-29a.


Sujet(s)
Cardiopathies congénitales/génétique , Hypertension pulmonaire/génétique , Hypertrophie ventriculaire droite/génétique , microARN/génétique , Transcriptome , Fonction ventriculaire droite/génétique , Remodelage ventriculaire/génétique , Adaptation physiologique , Animaux , Modèles animaux de maladie humaine , Fibrose , Analyse de profil d'expression de gènes/méthodes , Cardiopathies congénitales/métabolisme , Cardiopathies congénitales/physiopathologie , Ventricules cardiaques/métabolisme , Ventricules cardiaques/physiopathologie , Hémodynamique , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/physiopathologie , Hypertrophie ventriculaire droite/métabolisme , Hypertrophie ventriculaire droite/physiopathologie , microARN/métabolisme , Ovis aries
16.
Cardiovasc Res ; 113(12): 1433-1440, 2017 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-28957533

RÉSUMÉ

Right ventricular (RV) remodelling is a lesser understood process of the chronic, progressive transformation of the RV structure leading to reduced functional capacity and subsequent failure. Besides conditions concerning whole hearts, some pathology selectively affects the RV, leading to a distinct RV-specific clinical phenotype. MicroRNAs have been identified as key regulators of biological processes that drive the progression of chronic diseases. The role of microRNAs in diseases affecting the left ventricle has been studied for many years, however there is still limited information on microRNAs specific to diseases in the right ventricle. Here, we review recently described details on the expression, regulation, and function of microRNAs in the pathological remodelling of the right heart. Recently identified strategies using microRNAs as pharmacological targets or biomarkers will be highlighted. Increasing knowledge of pathogenic microRNAs will finally help improve our understanding of underlying distinct mechanisms and help utilize novel targets or biomarkers to develop treatments for patients suffering from right heart diseases.


Sujet(s)
Défaillance cardiaque/génétique , Hypertrophie ventriculaire droite/génétique , microARN/génétique , Dysfonction ventriculaire droite/génétique , Fonction ventriculaire droite/génétique , Remodelage ventriculaire/génétique , Animaux , Dysplasie ventriculaire droite arythmogène/complications , Dysplasie ventriculaire droite arythmogène/génétique , Dysplasie ventriculaire droite arythmogène/physiopathologie , Évolution de la maladie , Régulation de l'expression des gènes , Marqueurs génétiques , Défaillance cardiaque/étiologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/physiopathologie , Humains , Hypertension pulmonaire/complications , Hypertension pulmonaire/génétique , Hypertension pulmonaire/physiopathologie , Hypertrophie ventriculaire droite/étiologie , Hypertrophie ventriculaire droite/métabolisme , Hypertrophie ventriculaire droite/physiopathologie , microARN/métabolisme , Dysfonction ventriculaire droite/étiologie , Dysfonction ventriculaire droite/métabolisme , Dysfonction ventriculaire droite/physiopathologie
17.
Cardiovasc Res ; 113(12): 1521-1531, 2017 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-28957532

RÉSUMÉ

Arrhythmogenic cardiomyopathy (ACM) is a rare, heritable heart disease characterized by fibro-fatty replacement of the myocardium and a high degree of electric instability. It was first thought to be a congenital disorder, but is now regarded as a dystrophic heart muscle disease that develops over time. There is no curative treatment and current treatment strategies focus on attenuating the symptoms, slowing disease progression, and preventing life-threatening arrhythmias and sudden cardiac death. Identification of mutations in genes encoding desmosomal proteins and in other genes has led to insights into the disease pathogenesis and greatly facilitated identification of family members at risk. The disease phenotype is, however, highly variable and characterized by incomplete penetrance. Although the reasons are still poorly understood, sex, endurance exercise and a gene-dosage effect seem to play a role in these phenomena. The discovery of the genes and mutations implicated in ACM has allowed animal and cellular models to be generated, enabling researchers to start unravelling it's underlying molecular mechanisms. Observations in humans and in animal models suggest that reduced cell-cell adhesion affects gap junction and ion channel remodelling at the intercalated disc, and along with impaired desmosomal function, these can lead to perturbations in signalling cascades like the Wnt/ß-catenin and Hippo/YAP pathways. Perturbations of these pathways are also thought to lead to fibro-fatty replacement. A better understanding of the molecular processes may lead to new therapies that target specific pathways involved in ACM.


Sujet(s)
Dysplasie ventriculaire droite arythmogène , Mort subite cardiaque/étiologie , Mutation , Fonction ventriculaire gauche , Fonction ventriculaire droite , Remodelage ventriculaire , Animaux , Dysplasie ventriculaire droite arythmogène/diagnostic , Dysplasie ventriculaire droite arythmogène/génétique , Dysplasie ventriculaire droite arythmogène/mortalité , Dysplasie ventriculaire droite arythmogène/physiopathologie , Modèles animaux de maladie humaine , Prédisposition génétique à une maladie , Humains , Phénotype , Facteurs de risque , , Fonction ventriculaire gauche/génétique , Fonction ventriculaire droite/génétique , Remodelage ventriculaire/génétique
18.
Proc Natl Acad Sci U S A ; 113(31): 8801-6, 2016 08 02.
Article de Anglais | MEDLINE | ID: mdl-27432976

RÉSUMÉ

Hypoxic pulmonary vasoconstriction is correlated with pulmonary vascular remodeling. The hypoxia-inducible transcription factors (HIFs) HIF-1α and HIF-2α are known to contribute to the process of hypoxic pulmonary vascular remodeling; however, the specific role of pulmonary endothelial HIF expression in this process, and in the physiological process of vasoconstriction in response to hypoxia, remains unclear. Here we show that pulmonary endothelial HIF-2α is a critical regulator of hypoxia-induced pulmonary arterial hypertension. The rise in right ventricular systolic pressure (RVSP) normally observed following chronic hypoxic exposure was absent in mice with pulmonary endothelial HIF-2α deletion. The RVSP of mice lacking HIF-2α in pulmonary endothelium after exposure to hypoxia was not significantly different from normoxic WT mice and much lower than the RVSP values seen in WT littermate controls and mice with pulmonary endothelial deletion of HIF-1α exposed to hypoxia. Endothelial HIF-2α deletion also protected mice from hypoxia remodeling. Pulmonary endothelial deletion of arginase-1, a downstream target of HIF-2α, likewise attenuated many of the pathophysiological symptoms associated with hypoxic pulmonary hypertension. We propose a mechanism whereby chronic hypoxia enhances HIF-2α stability, which causes increased arginase expression and dysregulates normal vascular NO homeostasis. These data offer new insight into the role of pulmonary endothelial HIF-2α in regulating the pulmonary vascular response to hypoxia.


Sujet(s)
Arginase/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Endothélium vasculaire/métabolisme , Hypertension pulmonaire/métabolisme , Animaux , Arginase/génétique , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Hypoxie cellulaire , Cellules cultivées , Endothélium vasculaire/cytologie , Humains , Hypertension pulmonaire/génétique , Hypoxie , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Mâle , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Monoxyde d'azote/métabolisme , Fonction ventriculaire droite/génétique , Fonction ventriculaire droite/physiologie , Pression ventriculaire/génétique , Pression ventriculaire/physiologie
19.
Circulation ; 133(18): 1747-60, 2016 May 03.
Article de Anglais | MEDLINE | ID: mdl-26984938

RÉSUMÉ

BACKGROUND: The effect of a mutation in the bone morphogenetic protein receptor 2 (BMPR2) gene on right ventricular (RV) pressure overload in patients with pulmonary arterial hypertension is unknown. Therefore, we investigated RV function in patients who have pulmonary arterial hypertension with and without the BMPR2 mutation by combining in vivo measurements with molecular and histological analysis of human RV and left ventricular tissue. METHODS AND RESULTS: In total, 95 patients with idiopathic or familial pulmonary arterial hypertension were genetically screened for the presence of a BMPR2 mutation: 28 patients had a BMPR2 mutation, and 67 patients did not have a BMPR2 mutation. In vivo measurements were assessed using right heart catheterization and cardiac MRI. Despite a similar mean pulmonary artery pressure (noncarriers 54±15 versus mutation carriers 55±9 mm Hg) and pulmonary vascular resistance (755 [483-1043] versus 931 [624-1311] dynes·s(-1)·cm(-5)), mutation carriers presented with a more severely compromised RV function (RV ejection fraction: 37.6±12.8% versus 29.0±9%: P<0.05; cardiac index 2.7±0.9 versus 2.2±0.4 L·min(-1)·m(-2)). Differences continued to exist after treatment. To investigate the role of transforming growth factor ß and bone morphogenetic protein receptor II signaling, human RV and left ventricular tissue were studied in controls (n=6), mutation carriers (n=5), and noncarriers (n=11). However, transforming growth factor ß and bone morphogenetic protein receptor II signaling, and hypertrophy, apoptosis, fibrosis, capillary density, inflammation, and cardiac metabolism, as well, were similar between mutation carriers and noncarriers. CONCLUSIONS: Despite a similar afterload, RV function is more severely affected in mutation carriers than in noncarriers. However, these differences cannot be explained by a differential transforming growth factor ß, bone morphogenetic protein receptor II signaling, or cardiac adaptation.


Sujet(s)
Récepteurs de la protéine morphogénique osseuse de type II/génétique , Hypertension pulmonaire/génétique , Mutation/génétique , Dysfonction ventriculaire droite/génétique , Fonction ventriculaire droite/génétique , Adulte , Sujet âgé , Femelle , Humains , Hypertension pulmonaire/diagnostic , Mâle , Adulte d'âge moyen , Études rétrospectives , Dysfonction ventriculaire droite/diagnostic
20.
Eur Respir J ; 47(2): 553-63, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26647441

RÉSUMÉ

Sex hormones are linked to right ventricular (RV) function, but the relationship between genetic variation in these pathways and RV function is unknown.We performed a cross-sectional study of 2761 genotyped adults without cardiovascular disease. The relationships between RV measures and single nucleotide polymorphisms (SNPs) in 10 candidate genes were assessed. Urinary oestradiol (E2) metabolites produced by cytochrome P4501B1 (CYP1B1) and serum testosterone were measured in women and men respectively.In African-American (AA) women, the CYP1B1 SNP rs162561 was associated with RV ejection fraction (RVEF), such that each copy of the A allele was associated with a 2.0% increase in RVEF. Haplotype analysis revealed associations with RVEF in AA (global p<7.2×10(-6)) and white (global p=0.05) women. In white subjects, higher E2 metabolite levels were associated with significantly higher RVEF. In men, androgen receptors SNPs (rs1337080; rs5918764) were significantly associated with all RV measures and modified the relationship between testosterone and RVEF.Genetic variation in E2 metabolism and androgen signalling was associated with RV morphology in a sex-specific manner. The CYP1B1 SNP identified is in tight linkage disequilibrium with SNPs associated with pulmonary hypertension and oncogenesis, suggesting these pathways may underpin sexual dimorphism in RV failure.


Sujet(s)
Oestradiol/métabolisme , Récepteurs aux androgènes/génétique , Débit systolique/génétique , Dysfonction ventriculaire droite/génétique , Fonction ventriculaire droite/génétique , /génétique , Sujet âgé , Sujet âgé de 80 ans ou plus , Études de cohortes , Études transversales , Cytochrome P-450 CYP1B1/génétique , Femelle , Génotype , Haplotypes , Humains , Mâle , Adulte d'âge moyen , Polymorphisme de nucléotide simple , Études prospectives , Facteurs sexuels , /génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...