Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.584
Filtrer
1.
Cell Mol Life Sci ; 81(1): 258, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38878112

RÉSUMÉ

Pulmonary hypertension (PAH) is a cardiopulmonary disease in which pulmonary artery pressure continues to rise, leading to right heart failure and death. Otud6b is a member of the ubiquitin family and is involved in cell proliferation, apoptosis and inflammation. The aim of this study was to understand the role and mechanism of Otud6b in PAH. C57BL/6 and Calpain-1 knockout (KO) mice were exposed to a PAH model induced by 10% oxygen. Human pulmonary artery endothelial cells (HPACEs) and human pulmonary artery smooth muscle cells (HPASMCs) were exposed to 3% oxygen to establish an in vitro model. Proteomics was used to determine the role of Otud6b and its relationship to Calpain-1/HIF-1α signaling. The increased expression of Otud6b is associated with the progression of PAH. ROtud6b activates Otud6b, induces HIF-1α activation, increases the production of ET-1 and VEGF, and further aggravates endothelial injury. Reducing Otud6b expression by tracheal infusion of siOtud6b has the opposite effect, improving hemodynamic and cardiac response to PAH, reducing the release of Calpain-1 and HIF-1α, and eliminating the pro-inflammatory and apoptotic effects of Otud6b. At the same time, we also found that blocking Calpain-1 reduced the effect of Otud6b on HIF-1α, and inhibiting HIF-1α reduced the expression of Calpain-1 and Otud6b. Our study shows that increased Otud6b expression during hypoxia promotes the development of PAH models through a positive feedback loop between HIF-1α and Calpain-1. Therefore, we use Otud6b as a biomarker of PAH severity, and regulating Otud6b expression may be an effective target for the treatment of PAH.


Sujet(s)
Calpain , Sous-unité alpha du facteur-1 induit par l'hypoxie , Souris de lignée C57BL , Souris knockout , Transduction du signal , Animaux , Humains , Mâle , Souris , Calpain/métabolisme , Calpain/génétique , Modèles animaux de maladie humaine , Endopeptidases/métabolisme , Endopeptidases/génétique , Cellules endothéliales/métabolisme , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Hypertension artérielle pulmonaire/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie
2.
Cell Mol Life Sci ; 81(1): 256, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38866991

RÉSUMÉ

Pulmonary hypertension (PH) is characterized by vascular remodeling predominantly driven by a phenotypic switching in pulmonary artery smooth muscle cells (PASMCs). However, the underlying mechanisms for this phenotypic alteration remain incompletely understood. Here, we identified that RNA methyltransferase METTL3 is significantly elevated in the lungs of hypoxic PH (HPH) mice and rats, as well as in the pulmonary arteries (PAs) of HPH rats. Targeted deletion of Mettl3 in smooth muscle cells exacerbated hemodynamic consequences of hypoxia-induced PH and accelerated pulmonary vascular remodeling in vivo. Additionally, the absence of METTL3 markedly induced phenotypic switching in PASMCs in vitro. Mechanistically, METTL3 depletion attenuated m6A modification and hindered the processing of pri-miR-143/145, leading to a downregulation of miR-143-3p and miR-145-5p. Inhibition of hnRNPA2B1, an m6A mediator involved in miRNA maturation, similarly resulted in a significant reduction of miR-143-3p and miR-145-5p. We demonstrated that miR-145-5p targets Krüppel-like factor 4 (KLF4) and miR-143-3p targets fascin actin-bundling protein 1 (FSCN1) in PASMCs. The decrease of miR-145-5p subsequently induced an upregulation of KLF4, which in turn suppressed miR-143/145 transcription, establishing a positive feedback circuit between KLF4 and miR-143/145. This regulatory circuit facilitates the persistent suppression of contractile marker genes, thereby sustaining PASMC phenotypic switch. Collectively, hypoxia-induced upregulation of METTL3, along with m6A mediated regulation of miR-143/145, might serve as a protective mechanism against phenotypic switch of PASMCs. Our results highlight a potential therapeutic strategy targeting m6A modified miR-143/145-KLF4 loop in the treatment of PH.


Sujet(s)
Adénosine , Facteur-4 de type Kruppel , Facteurs de transcription Krüppel-like , Methyltransferases , microARN , Myocytes du muscle lisse , Artère pulmonaire , Facteur-4 de type Kruppel/métabolisme , Animaux , microARN/génétique , microARN/métabolisme , Artère pulmonaire/métabolisme , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Myocytes du muscle lisse/métabolisme , Souris , Adénosine/analogues et dérivés , Adénosine/métabolisme , Methyltransferases/métabolisme , Methyltransferases/génétique , Rats , Phénotype , Mâle , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/anatomopathologie , Muscles lisses vasculaires/métabolisme , Souris de lignée C57BL , Remodelage vasculaire/génétique , Rat Sprague-Dawley , Humains
3.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892440

RÉSUMÉ

NOTCH3 receptor signaling has been linked to the regulation of smooth muscle cell proliferation and the maintenance of smooth muscle cells in an undifferentiated state. Pulmonary arterial hypertension (World Health Organization Group 1 idiopathic disease: PAH) is a fatal disease characterized clinically by elevated pulmonary vascular resistance caused by extensive vascular smooth muscle cell proliferation, perivascular inflammation, and asymmetric neointimal hyperplasia in precapillary pulmonary arteries. In this review, a detailed overview of the specific role of NOTCH3 signaling in PAH, including its mechanisms of activation by a select ligand, downstream signaling effectors, and physiologic effects within the pulmonary vascular tree, is provided. Animal models showing the importance of the NOTCH3 pathway in clinical PAH will be discussed. New drugs and biologics that inhibit NOTCH3 signaling and reverse this deadly disease are highlighted.


Sujet(s)
Hypertension artérielle pulmonaire , Récepteur Notch3 , Transduction du signal , Humains , Récepteur Notch3/métabolisme , Récepteur Notch3/génétique , Animaux , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie
4.
Sci Rep ; 14(1): 14056, 2024 06 18.
Article de Anglais | MEDLINE | ID: mdl-38890390

RÉSUMÉ

Pulmonary arterial hypertension (PAH) is a progressive disease characterized by pulmonary vascular remolding and occlusion, leading to the elevated pulmonary arterial pressures, right ventricular hypertrophy, and eventual heart failure if left untreated. Understanding the molecular mechanisms underlying the development and progression of pulmonary hypertension (PH) is crucial for devising efficient therapeutic approaches for the disease. Lung homogenates were collected weekly and underwent RNA-sequencing in the monocrotaline (MCT)-induced PH rat model to explore genes associated with PH progression. Statistical analyses revealed 1038, 1244, and 3125 significantly altered genes (P < 0.05, abs (log2fold change) > log21.5) between control and MCT-exposed rats during the first, second, and third week, respectively. Pathway enrichment analyses revealed involvement of cell cycle and innate immune system for the upregulated genes, GPCR and VEGF signaling for the downregulated genes. Furthermore, qRT-PCR validated upregulation of representative genes associated with cell cycle including Cdc25c (cell division cycle 25C), Cdc45, Top2a (topoisomerase IIα), Ccna2 (cyclin A2) and Ccnb1 (cyclin B1). Western blot and immunofluorescence analysis confirmed increases in PCNA, Ccna2, Top2a, along with other proliferation markers in the lung tissue of MCT-treated rats. In summary, RNA sequencing data highlights the significance of cell proliferation in progression of rodent PH.


Sujet(s)
Prolifération cellulaire , Modèles animaux de maladie humaine , Évolution de la maladie , Analyse de profil d'expression de gènes , Hypertension pulmonaire , Monocrotaline , Animaux , Rats , Hypertension pulmonaire/induit chimiquement , Hypertension pulmonaire/génétique , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Mâle , Rat Sprague-Dawley , Transcriptome , Poumon/anatomopathologie , Poumon/métabolisme
5.
Cells ; 13(11)2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38891046

RÉSUMÉ

Fibroblasts, among the most prevalent and widely distributed cell types in the human body, play a crucial role in defining tissue structure. They do this by depositing and remodeling extracellular matrixes and organizing functional tissue networks, which are essential for tissue homeostasis and various human diseases. Pulmonary hypertension (PH) is a devastating syndrome with high mortality, characterized by remodeling of the pulmonary vasculature and significant cellular and structural changes within the intima, media, and adventitia layers. Most research on PH has focused on alterations in the intima (endothelial cells) and media (smooth muscle cells). However, research over the past decade has provided strong evidence of the critical role played by pulmonary artery adventitial fibroblasts in PH. These fibroblasts exhibit the earliest, most dramatic, and most sustained proliferative, apoptosis-resistant, and inflammatory responses to vascular stress. This review examines the aberrant phenotypes of PH fibroblasts and their role in the pathogenesis of PH, discusses potential molecular signaling pathways underlying these activated phenotypes, and highlights areas of research that merit further study to identify promising targets for the prevention and treatment of PH.


Sujet(s)
Fibroblastes , Hypertension pulmonaire , Humains , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/physiopathologie , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Animaux , Transduction du signal , Artère pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme
6.
Commun Biol ; 7(1): 693, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844781

RÉSUMÉ

Pericyte dysfunction, with excessive migration, hyperproliferation, and differentiation into smooth muscle-like cells contributes to vascular remodeling in Pulmonary Arterial Hypertension (PAH). Augmented expression and action of growth factors trigger these pathological changes. Endogenous factors opposing such alterations are barely known. Here, we examine whether and how the endothelial hormone C-type natriuretic peptide (CNP), signaling through the cyclic guanosine monophosphate (cGMP) -producing guanylyl cyclase B (GC-B) receptor, attenuates the pericyte dysfunction observed in PAH. The results demonstrate that CNP/GC-B/cGMP signaling is preserved in lung pericytes from patients with PAH and prevents their growth factor-induced proliferation, migration, and transdifferentiation. The anti-proliferative effect of CNP is mediated by cGMP-dependent protein kinase I and inhibition of the Phosphoinositide 3-kinase (PI3K)/AKT pathway, ultimately leading to the nuclear stabilization and activation of the Forkhead Box O 3 (FoxO3) transcription factor. Augmentation of the CNP/GC-B/cGMP/FoxO3 signaling pathway might be a target for novel therapeutics in the field of PAH.


Sujet(s)
Prolifération cellulaire , GMP cyclique , Protéine O3 à motif en tête de fourche , Peptide natriurétique de type C , Péricytes , Transduction du signal , Humains , Péricytes/métabolisme , Péricytes/anatomopathologie , Peptide natriurétique de type C/métabolisme , GMP cyclique/métabolisme , Protéine O3 à motif en tête de fourche/métabolisme , Protéine O3 à motif en tête de fourche/génétique , Mâle , Femelle , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Adulte d'âge moyen , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Adulte , Récepteur facteur natriurétique auriculaire/métabolisme , Récepteur facteur natriurétique auriculaire/génétique , Cellules cultivées
7.
Open Vet J ; 14(5): 1216-1223, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38938438

RÉSUMÉ

Background: Pulmonary capillary hemangiomatosis (PCH) is an idiopathic disease with the anomalous proliferation of a small capillary-like vessel in the pulmonary tissue, which can lead to a severe form of PH. There are only several cases of PCH described in veterinary literature: 27 cases in dogs and 2 cases in cats. In veterinary medicine, PH is mostly recognized as a consequence of left heart failure as a progression of the postcapillary PH to the precapillary form. PCH is mostly described as a primary disease, but resistant postcapillary PH with the high possibility of pulmonary edema raises speculation that PCH could be a secondary malformation to the left heart disease. Aim: Discover the features associated with the shift between left- and right-sided heart disease in the context of PH development. Methods: Retrospective analysis of materials from cats and dogs with histological markers of PCH (sPCH) versus those with right heart failure (RHF). Results: Animals with histological and immunohistochemistry markers of PCH had a previous history of disease with left heart volume overload. There were no differences between the groups in radiography and gross pathology. Histologically, pulmonary fibrosis and arteriopathy could be found in RHF; in sPCH-a duplication of capillaries in alveolar septa and bizarre proliferation in surrounding structures. Conclusion: PCH could be a secondary pattern of vascular remodeling due to volume overload.


Sujet(s)
Maladies des chats , Maladies des chiens , Hypertension pulmonaire , Animaux , Chiens , Maladies des chats/anatomopathologie , Maladies des chats/diagnostic , Maladies des chiens/anatomopathologie , Maladies des chiens/diagnostic , Chats , Hypertension pulmonaire/médecine vétérinaire , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/étiologie , Études rétrospectives , Mâle , Femelle , Hémangiome capillaire/médecine vétérinaire , Hémangiome capillaire/anatomopathologie , Hémangiome capillaire/complications , Défaillance cardiaque/médecine vétérinaire , Défaillance cardiaque/étiologie , Défaillance cardiaque/anatomopathologie , Tumeurs du poumon/médecine vétérinaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/complications
8.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 233-237, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38836656

RÉSUMÉ

Nur77 is a member of the NR4A subfamily of orphan nuclear receptors that is expressed and has a function within the immune system. This study aimed to investigate the role of Nur77 in hypoxic pulmonary hypertension. SPF male SD rats were exposed in hypobaric chamber simulating 5000 m high altitude for 0, 3, 7, 14, 21 or 28 days. Rat pulmonary artery smooth muscle cells (RPASMCs) were cultured under normoxic conditions (5% CO2-95% ambient air) or hypoxic conditions (5% O2 for 6 h, 12 h, 24 h, 48 h). Hypoxic rats developed pulmonary arterial remodeling and right ventricular hypertrophy with significantly increased pulmonary arterial pressure. The levels of Nur77, HIF-1α and PNCA were upregulated in pulmonary arterial smooth muscle from hypoxic rats. Silencing of either Nur77 or HIF-1α attenuated hypoxia-induced proliferation. Silencing of HIF-1α down-regulated Nur77 protein level, but Nur77 silence did not reduce HIF-1α. Nur77 was not con-immunoprecipitated with HIF-1α. This study demonstrated that Nur77 acted as a downstream regulator of HIF-1α under hypoxia, and plays a critical role in the hypoxia-induced pulmonary vascular remodeling, which is regulated by HIF-1α. Nur77 maybe a novel target of HPH therapy.


Sujet(s)
Hypertension pulmonaire , Sous-unité alpha du facteur-1 induit par l'hypoxie , Hypoxie , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires , Artère pulmonaire , Rat Sprague-Dawley , Remodelage vasculaire , Animaux , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-1 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Remodelage vasculaire/génétique , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Mâle , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Hypoxie/métabolisme , Prolifération cellulaire , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Rats , Hypertrophie ventriculaire droite/métabolisme , Hypertrophie ventriculaire droite/anatomopathologie , Hypertrophie ventriculaire droite/physiopathologie , Hypertrophie ventriculaire droite/génétique , Cellules cultivées
9.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38892401

RÉSUMÉ

Increased mitochondrial reactive oxygen species (ROS) formation is important for the development of right ventricular (RV) hypertrophy (RVH) and failure (RVF) during pulmonary hypertension (PH). ROS molecules are produced in different compartments within the cell, with mitochondria known to produce the strongest ROS signal. Among ROS-forming mitochondrial proteins, outer-mitochondrial-membrane-located monoamine oxidases (MAOs, type A or B) are capable of degrading neurotransmitters, thereby producing large amounts of ROS. In mice, MAO-B is the dominant isoform, which is present in almost all cell types within the heart. We analyzed the effect of an inducible cardiomyocyte-specific knockout of MAO-B (cmMAO-B KO) for the development of RVH and RVF in mice. Right ventricular hypertrophy was induced by pulmonary artery banding (PAB). RV dimensions and function were measured through echocardiography. ROS production (dihydroethidium staining), protein kinase activity (PamStation device), and systemic hemodynamics (in vivo catheterization) were assessed. A significant decrease in ROS formation was measured in cmMAO-B KO mice during PAB compared to Cre-negative littermates, which was associated with reduced activity of protein kinases involved in hypertrophic growth. In contrast to littermates in which the RV was dilated and hypertrophied following PAB, RV dimensions were unaffected in response to PAB in cmMAO-B KO mice, and no decline in RV systolic function otherwise seen in littermates during PAB was measured in cmMAO-B KO mice. In conclusion, cmMAO-B KO mice are protected against RV dilatation, hypertrophy, and dysfunction following RV pressure overload compared to littermates. These results support the hypothesis that cmMAO-B is a key player in causing RV hypertrophy and failure during PH.


Sujet(s)
Hypertension pulmonaire , Hypertrophie ventriculaire droite , Souris knockout , Monoamine oxidase , Espèces réactives de l'oxygène , Animaux , Hypertrophie ventriculaire droite/métabolisme , Hypertrophie ventriculaire droite/génétique , Hypertrophie ventriculaire droite/étiologie , Hypertrophie ventriculaire droite/anatomopathologie , Monoamine oxidase/génétique , Monoamine oxidase/métabolisme , Monoamine oxidase/déficit , Hypertension pulmonaire/génétique , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Souris , Espèces réactives de l'oxygène/métabolisme , Myocytes cardiaques/métabolisme , Myocytes cardiaques/anatomopathologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/étiologie , Défaillance cardiaque/génétique , Défaillance cardiaque/anatomopathologie , Mâle , Modèles animaux de maladie humaine , Ventricules cardiaques/anatomopathologie , Ventricules cardiaques/métabolisme , Dysfonction ventriculaire droite/métabolisme , Dysfonction ventriculaire droite/génétique , Dysfonction ventriculaire droite/étiologie , Dysfonction ventriculaire droite/anatomopathologie
10.
Genes Genomics ; 46(7): 751-762, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38733520

RÉSUMÉ

BACKGROUND: The apoptosis-resistant pulmonary arterial endothelial cells (PAECs) are known to be major players in the pulmonary remodeling of pulmonary arterial hypertension (PAH) and exhibit an abnormal metabolic profile with mitochondrial dysfunction. Mitochondrial fission has been shown to regulate the apoptosis of several cell types, but this is largely unexplored in the PAECs. OBJECTIVE: The roles of mitochondrial fission control by Dynamin related protein-1 (DRP1) in the development of PAECs apoptosis suppression were investigated in present study and the potential mechanisms behind this were furtherly explored. METHODS: The mitochondrial morphology was investigated in PAECs from PAH rats with the pulmonary plexiform lesions, and the relations of it with DRP1 expression and apoptosis were furtherly identified in apoptosis-resistant PAECs induced by hypoxia. PAECs were isolated from rats with severe PAH and from normal subjects, the apoptotic-resistant PAECs were induced by hypoxia. DRP1 gene knockdown was achieved via DRP1-siRNA, DRP1 and STAT3 phosphorylation were blocked using its inhibitors, respectively. Apoptosis was analyzed by flow cytometry, and mitochondrial morphology was investigated by transmission electron microscope and confocal microscopy. RESULTS: The PAECs isolated from PAH rats with the pulmonary plexiform-like lesions and displayed lower apoptotic rate with increased DRP1 expression and mitochondrial fragmentation. In addition, similar observations were achieved in apoptosis-resistant PAECs induced by hypoxia. Targeting DRP1 using siRNA and pharmacologic blockade prevented the mitochondrial fission and subsequent apoptotic resistance in PAECs under hypoxia. Mechanistically, STAT3 phosphorylation at Tyr705 was shown to be activated in both PAH and hypoxia-treated PAECs, leading to the regulation of DRP1 expression. Of importance, targeting STAT3Tyr705 phosphorylation prevented DRP1 disruption on apoptosis in PAECs under hypoxia. CONCLUSIONS: These data indicated that STAT3 phosphorylation at Tyr705 impacted DRP1-controlled mitochondrial fission during the development of apoptosis-resistance in PAECs, suggesting mitochondrial dynamics may represent a therapeutic target for PAH.


Sujet(s)
Apoptose , Dynamines , Cellules endothéliales , Dynamique mitochondriale , Artère pulmonaire , Facteur de transcription STAT-3 , Animaux , Dynamines/métabolisme , Dynamines/génétique , Dynamique mitochondriale/génétique , Rats , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Cellules endothéliales/métabolisme , Phosphorylation , Artère pulmonaire/métabolisme , Artère pulmonaire/cytologie , Rat Sprague-Dawley , Mâle , Mitochondries/métabolisme , Mitochondries/génétique , Cellules cultivées , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/anatomopathologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/anatomopathologie
11.
Int J Mol Sci ; 25(10)2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38791441

RÉSUMÉ

Pulmonary arterial hypertension (PAH) is a progressive cardiopulmonary disease characterized by pathologic vascular remodeling of small pulmonary arteries. Endothelial dysfunction in advanced PAH is associated with proliferation, apoptosis resistance, and endothelial to mesenchymal transition (EndoMT) due to aberrant signaling. DLL4, a cell membrane associated NOTCH ligand, plays a pivotal role maintaining vascular integrity. Inhibition of DLL4 has been associated with the development of pulmonary hypertension, but the mechanism is incompletely understood. Here we report that BMPR2 silencing in pulmonary artery endothelial cells (PAECs) activated AKT and suppressed the expression of DLL4. Consistent with these in vitro findings, increased AKT activation and reduced DLL4 expression was found in the small pulmonary arteries of patients with PAH. Increased NOTCH1 activation through exogenous DLL4 blocked AKT activation, decreased proliferation and reversed EndoMT. Exogenous and overexpression of DLL4 induced BMPR2 and PPRE promoter activity, and BMPR2 and PPARG mRNA in idiopathic PAH (IPAH) ECs. PPARγ, a nuclear receptor associated with EC homeostasis, suppressed by BMPR2 loss was induced and activated by DLL4/NOTCH1 signaling in both BMPR2-silenced and IPAH ECs, reversing aberrant phenotypic changes, in part through AKT inhibition. Directly blocking AKT or restoring DLL4/NOTCH1/PPARγ signaling may be beneficial in preventing or reversing the pathologic vascular remodeling of PAH.


Sujet(s)
Récepteurs de la protéine morphogénique osseuse de type II , Cellules endothéliales , Récepteur PPAR gamma , Protéines proto-oncogènes c-akt , Artère pulmonaire , Récepteur Notch1 , Transduction du signal , Humains , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Récepteurs de la protéine morphogénique osseuse de type II/génétique , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Récepteur Notch1/métabolisme , Récepteur Notch1/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Cellules endothéliales/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines de liaison au calcium/métabolisme , Protéines de liaison au calcium/génétique , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/anatomopathologie , Mâle , Prolifération cellulaire , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/anatomopathologie , Femelle , Cellules cultivées
12.
Circ Res ; 135(1): 93-109, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38770649

RÉSUMÉ

BACKGROUND: Hyperproliferation of pulmonary arterial smooth muscle cells (PASMCs) and consequent pulmonary vascular remodeling are the crucial pathological features of pulmonary hypertension (PH). Protein methylation has been shown to be critically involved in PASMC proliferation and PH, but the underlying mechanism remains largely unknown. METHODS: PH animal models were generated by treating mice/rats with chronic hypoxia for 4 weeks. SMYD2-vTg mice (vascular smooth muscle cell-specific suppressor of variegation, enhancer of zeste, trithorax and myeloid Nervy DEAF-1 (deformed epidural auto-regulatory factor-1) domain-containing protein 2 transgenic) or wild-type rats and mice treated with LLY-507 (3-cyano-5-{2-[4-[2-(3-methylindol-1-yl)ethyl]piperazin-1-yl]-phenyl}-N-[(3-pyrrolidin-1-yl)propyl]benzamide) were used to investigate the function of SMYD2 (suppressor of variegation, enhancer of zeste, trithorax and myeloid Nervy DEAF-1 domain-containing protein 2) on PH development in vivo. Primary cultured rat PASMCs with SMYD2 knockdown or overexpression were used to explore the effects of SMYD2 on proliferation and to decipher the underlying mechanism. RESULTS: We demonstrated that the expression of the lysine methyltransferase SMYD2 was upregulated in the smooth muscle cells of pulmonary arteries from patients with PH and hypoxia-exposed rats/mice and in the cytoplasm of hypoxia-induced rat PASMCs. More importantly, targeted inhibition of SMYD2 by LLY-507 significantly attenuated hypoxia-induced pulmonary vascular remodeling and PH development in both male and female rats in vivo and reduced rat PASMC hyperproliferation in vitro. In contrast, SMYD2-vTg mice exhibited more severe PH phenotypes and related pathological changes than nontransgenic mice after 4 weeks of chronic hypoxia treatment. Furthermore, SMYD2 overexpression promoted, while SMYD2 knockdown suppressed, the proliferation of rat PASMCs by affecting the cell cycle checkpoint between S and G2 phases. Mechanistically, we revealed that SMYD2 directly interacted with and monomethylated PPARγ (peroxisome proliferator-activated receptor gamma) to inhibit the nuclear translocation and transcriptional activity of PPARγ, which further promoted mitophagy to facilitate PASMC proliferation and PH development. Furthermore, rosiglitazone, a PPARγ agonist, largely abolished the detrimental effects of SMYD2 overexpression on PASMC proliferation and PH. CONCLUSIONS: Our results demonstrated that SMYD2 monomethylates nonhistone PPARγ and inhibits its nuclear translocation and activation to accelerate PASMC proliferation and PH by triggering mitophagy, indicating that targeting SMYD2 or activating PPARγ are potential strategies for the prevention of PH.


Sujet(s)
Histone-lysine N-methyltransferase , Hypertension pulmonaire , Hypoxie , Mitophagie , Muscles lisses vasculaires , Myocytes du muscle lisse , Récepteur PPAR gamma , Artère pulmonaire , Rat Sprague-Dawley , Animaux , Récepteur PPAR gamma/métabolisme , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/génétique , Hypoxie/complications , Hypoxie/métabolisme , Souris , Rats , Mâle , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Histone-lysine N-methyltransferase/métabolisme , Histone-lysine N-methyltransferase/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Artère pulmonaire/anatomopathologie , Artère pulmonaire/métabolisme , Souris transgéniques , Cellules cultivées , Prolifération cellulaire , Remodelage vasculaire , Humains , Souris de lignée C57BL , Méthylation
13.
Circ Res ; 135(1): 60-75, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38770652

RÉSUMÉ

BACKGROUND: Pathogenic concepts of right ventricular (RV) failure in pulmonary arterial hypertension focus on a critical loss of microvasculature. However, the methods underpinning prior studies did not take into account the 3-dimensional (3D) aspects of cardiac tissue, making accurate quantification difficult. We applied deep-tissue imaging to the pressure-overloaded RV to uncover the 3D properties of the microvascular network and determine whether deficient microvascular adaptation contributes to RV failure. METHODS: Heart sections measuring 250-µm-thick were obtained from mice after pulmonary artery banding (PAB) or debanding PAB surgery and properties of the RV microvascular network were assessed using 3D imaging and quantification. Human heart tissues harvested at the time of transplantation from pulmonary arterial hypertension cases were compared with tissues from control cases with normal RV function. RESULTS: Longitudinal 3D assessment of PAB mouse hearts uncovered complex microvascular remodeling characterized by tortuous, shorter, thicker, highly branched vessels, and overall preserved microvascular density. This remodeling process was reversible in debanding PAB mice in which the RV function recovers over time. The remodeled microvasculature tightly wrapped around the hypertrophied cardiomyocytes to maintain a stable contact surface to cardiomyocytes as an adaptation to RV pressure overload, even in end-stage RV failure. However, microvasculature-cardiomyocyte contact was impaired in areas with interstitial fibrosis where cardiomyocytes displayed signs of hypoxia. Similar to PAB animals, microvascular density in the RV was preserved in patients with end-stage pulmonary arterial hypertension, and microvascular architectural changes appeared to vary by etiology, with patients with pulmonary veno-occlusive disease displaying a lack of microvascular complexity with uniformly short segments. CONCLUSIONS: 3D deep tissue imaging of the failing RV in PAB mice, pulmonary hypertension rats, and patients with pulmonary arterial hypertension reveals complex microvascular changes to preserve the microvascular density and maintain a stable microvascular-cardiomyocyte contact. Our studies provide a novel framework to understand microvascular adaptation in the pressure-overloaded RV that focuses on cell-cell interaction and goes beyond the concept of capillary rarefaction.


Sujet(s)
Hypertension pulmonaire , Imagerie tridimensionnelle , Souris de lignée C57BL , Animaux , Humains , Souris , Hypertension pulmonaire/physiopathologie , Hypertension pulmonaire/imagerie diagnostique , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/anatomopathologie , Mâle , Ventricules cardiaques/physiopathologie , Ventricules cardiaques/imagerie diagnostique , Ventricules cardiaques/anatomopathologie , Microvaisseaux/physiopathologie , Microvaisseaux/imagerie diagnostique , Microvaisseaux/anatomopathologie , Remodelage vasculaire , Artère pulmonaire/physiopathologie , Artère pulmonaire/imagerie diagnostique , Artère pulmonaire/anatomopathologie , Dysfonction ventriculaire droite/physiopathologie , Dysfonction ventriculaire droite/étiologie , Dysfonction ventriculaire droite/imagerie diagnostique , Fonction ventriculaire droite , Remodelage ventriculaire , Modèles animaux de maladie humaine , Myocytes cardiaques/anatomopathologie
14.
Biochem Biophys Res Commun ; 723: 150159, 2024 Sep 03.
Article de Anglais | MEDLINE | ID: mdl-38815488

RÉSUMÉ

Exercise has been recognized as an effective intervention in the treatment of pulmonary arterial hypertension (PAH), supported by numerous studies. However, the precise effects of exercise on pulmonary function remain to be fully elucidated. In this study, using a rat model of swimming exercise training and monocrotaline-induced PAH, we aimed to explore its impact on pulmonary morphology and function. Our investigations revealed that MCT-treated rats exhibited augmented mean pulmonary arterial pressure (MPAP) and pulmonary vascular remodeling, which can be attenuated by 4 weeks of swimming exercise training (60 min/day, 5 days/week). Notably, MCT-treated rats showed impaired pulmonary function, as manifested by decreased tidal volume and dynamic compliance, which were reversed by exercise training. Assessment of pulmonary substrate in PAH rats indicated a prominent pro-inflammatory substrate, evidenced by macrophage accumulation through quantitative immunohistological analysis of macrophage-like cell expression (CD68), and extracellular matrix remodeling, evaluated by Masson staining. Importantly, both the pro-inflammatory substrate and extracellular matrix remodeling were ameliorated by swimming exercise training. Additionally, serum biochemical analysis demonstrated elevated levels of low-density lipoprotein cholesterol and Apolipoprotein B following MCT treatment, which were reduced with exercise intervention. Moreover, exercise enhanced systemic insulin sensitivity in both MCT-treated and untreated rats. Notably, MCT and exercise treatment both decreased fasting blood glucose (FBG) levels in rats, whereas exercise training reinstated FBG levels to normal in MCT-treated rats. In summary, our study suggests that swimming exercise confers a pulmonary protective effect in MCT-induced PAH rats, highlighting the potential importance of exercise-based rehabilitation in the management of PAH.


Sujet(s)
Hypertension pulmonaire , Insulinorésistance , Monocrotaline , Conditionnement physique d'animal , Rat Sprague-Dawley , Natation , Animaux , Monocrotaline/toxicité , Mâle , Rats , Hypertension pulmonaire/induit chimiquement , Hypertension pulmonaire/thérapie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Hypertension pulmonaire/physiopathologie , Poumon/anatomopathologie , Poumon/métabolisme , Remodelage vasculaire
15.
Int J Mol Sci ; 25(9)2024 Apr 26.
Article de Anglais | MEDLINE | ID: mdl-38731946

RÉSUMÉ

Systemic sclerosis (SSc) is a heterogeneous disease characterized by autoimmunity, vasculopathy, and fibrosis which affects the skin and internal organs. One key aspect of SSc vasculopathy is pulmonary arterial hypertension (SSc-PAH) which represents a leading cause of morbidity and mortality in patients with SSc. The pathogenesis of pulmonary hypertension is complex, with multiple vascular cell types, inflammation, and intracellular signaling pathways contributing to vascular pathology and remodeling. In this review, we focus on shared molecular features of pulmonary hypertension and those which make SSc-PAH a unique entity. We highlight advances in the understanding of the clinical and translational science pertinent to this disease. We first review clinical presentations and phenotypes, pathology, and novel biomarkers, and then highlight relevant animal models, key cellular and molecular pathways in pathogenesis, and explore emerging treatment strategies in SSc-PAH.


Sujet(s)
Hypertension artérielle pulmonaire , Sclérodermie systémique , Humains , Sclérodermie systémique/complications , Sclérodermie systémique/anatomopathologie , Animaux , Hypertension artérielle pulmonaire/étiologie , Hypertension artérielle pulmonaire/métabolisme , Marqueurs biologiques , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , , Transduction du signal
16.
Cell Metab ; 36(6): 1335-1350.e8, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38701775

RÉSUMÉ

Perivascular collagen deposition by activated fibroblasts promotes vascular stiffening and drives cardiovascular diseases such as pulmonary hypertension (PH). Whether and how vascular fibroblasts rewire their metabolism to sustain collagen biosynthesis remains unknown. Here, we found that inflammation, hypoxia, and mechanical stress converge on activating the transcriptional coactivators YAP and TAZ (WWTR1) in pulmonary arterial adventitial fibroblasts (PAAFs). Consequently, YAP and TAZ drive glutamine and serine catabolism to sustain proline and glycine anabolism and promote collagen biosynthesis. Pharmacologic or dietary intervention on proline and glycine anabolic demand decreases vascular stiffening and improves cardiovascular function in PH rodent models. By identifying the limiting metabolic pathways for vascular collagen biosynthesis, our findings provide guidance for incorporating metabolic and dietary interventions for treating cardiopulmonary vascular disease.


Sujet(s)
Glutamine , Sérine , Rigidité vasculaire , Animaux , Glutamine/métabolisme , Sérine/métabolisme , Mâle , Souris , Souris de lignée C57BL , Fibroblastes/métabolisme , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie , Humains , Collagène/métabolisme , Rats
17.
Clin Respir J ; 18(5): e13771, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38747117

RÉSUMÉ

BACKGROUND: Hypertension is a main contributing factor of cardiovascular diseases; deregulated circular RNAs are involved in the pathogenesis of pulmonary arterial hypertension (PAH). Herein, we evaluated the function and mechanism of circST6GAL1 in PAH process. METHODS: Human pulmonary artery smooth muscle cells (HPASMCs) were cultured in hypoxic environment for functional analysis. The cell counting kit-8, 5-ethynyl-2'-deoxyuridine, wound healing, and flow cytometry assays were used to investigate cell proliferation, migration, and apoptosis. qRT-PCR and Western blotting analyses were used for level measurement of genes and proteins. The binding between miR-509-5p and circST6GAL1 or multiple C2 and transmembrane domain containing 2 (MCTP2) was analyzed by dual-luciferase reporter, RNA immunoprecipitation, and pull-down assays. The monocrotaline (MCT)-induced PAH mouse models were established for in vivo assay. RESULTS: CircST6GAL1 was highly expressed in PAH patients and hypoxia-induced HPASMCs. Functionally, circST6GAL1 deficiency reversed hypoxia-induced proliferation and migration, as well as apoptosis arrest in HPASMCs. Mechanistically, circST6GAL1 directly targeted miR-509-5p, and MCTP2 was a target of miR-509-5p. Rescue assays showed that the regulatory effects of circST6GAL1 deficiency on hypoxia-induced HPASMCs were abolished. Moreover, forced expression of miR-509-5p suppressed HPASMC proliferation and migration and induced cell apoptosis under hypoxia stimulation, while these effects were abolished by MCTP2 overexpression. Moreover, circST6GAL1 silencing improved MCT-induced pulmonary vascular remodeling and PAH. CONCLUSION: CircST6GAL1 deficiency reversed hypoxia-induced proliferation and migration, as well as apoptosis arrest in HPASMCs, and alleviated pulmonary vascular remodeling in MCT-induced PAH mouse models through the miR-509-5p/MCTP2 axis, indicating a potential therapeutic target for PAH.


Sujet(s)
Apoptose , Prolifération cellulaire , microARN , Hypertension artérielle pulmonaire , ARN circulaire , Humains , microARN/génétique , microARN/métabolisme , Souris , Animaux , ARN circulaire/génétique , ARN circulaire/métabolisme , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , Myocytes du muscle lisse/métabolisme , Mâle , Mouvement cellulaire/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Cellules cultivées , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/anatomopathologie
18.
Front Immunol ; 15: 1372957, 2024.
Article de Anglais | MEDLINE | ID: mdl-38779688

RÉSUMÉ

Background: Schistosomiasis is a common cause of pulmonary hypertension (PH) worldwide. Type 2 inflammation contributes to the development of Schistosoma-induced PH. Specifically, interstitial macrophages (IMs) derived from monocytes play a pivotal role by producing thrombospondin-1 (TSP-1), which in turn activates TGF-ß, thereby driving the pathology of PH. Resident and recruited IM subpopulations have recently been identified. We hypothesized that in Schistosoma-PH, one IM subpopulation expresses monocyte recruitment factors, whereas recruited monocytes become a separate IM subpopulation that expresses TSP-1. Methods: Mice were intraperitoneally sensitized and then intravenously challenged with S. mansoni eggs. Flow cytometry on lungs and blood was performed on wildtype and reporter mice to identify IM subpopulations and protein expression. Single-cell RNA sequencing (scRNAseq) was performed on flow-sorted IMs from unexposed and at day 1, 3 and 7 following Schistosoma exposure to complement flow cytometry based IM characterization and identify gene expression. Results: Flow cytometry and scRNAseq both identified 3 IM subpopulations, characterized by CCR2, MHCII, and FOLR2 expression. Following Schistosoma exposure, the CCR2+ IM subpopulation expanded, suggestive of circulating monocyte recruitment. Schistosoma exposure caused increased monocyte-recruitment ligand CCL2 expression in the resident FOLR2+ IM subpopulation. In contrast, the vascular pathology-driving protein TSP-1 was greatest in the CCR2+ IM subpopulation. Conclusion: Schistosoma-induced PH involves crosstalk between IM subpopulations, with increased expression of monocyte recruitment ligands by resident FOLR2+ IMs, and the recruitment of CCR2+ IMs which express TSP-1 that activates TGF-ß and causes PH.


Sujet(s)
Hypertension pulmonaire , Macrophages , Animaux , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/parasitologie , Hypertension pulmonaire/immunologie , Hypertension pulmonaire/anatomopathologie , Souris , Macrophages/immunologie , Macrophages/parasitologie , Phénotype , Schistosoma mansoni/immunologie , Souris de lignée C57BL , Schistosomiase/immunologie , Schistosomiase/complications , Schistosomiase/parasitologie , Modèles animaux de maladie humaine , Schistosomiase à Schistosoma mansoni/immunologie , Schistosomiase à Schistosoma mansoni/parasitologie , Schistosomiase à Schistosoma mansoni/complications , Schistosomiase à Schistosoma mansoni/anatomopathologie , Thrombospondine-1/génétique , Thrombospondine-1/métabolisme , Monocytes/immunologie , Récepteurs CCR2/génétique , Récepteurs CCR2/métabolisme , Femelle , Schistosoma/immunologie , Schistosoma/physiologie , Poumon/immunologie , Poumon/parasitologie , Poumon/anatomopathologie
19.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38732160

RÉSUMÉ

Despite the end of the pandemic, coronavirus disease 2019 (COVID-19) remains a major public health concern. The first waves of the virus led to a better understanding of its pathogenesis, highlighting the fact that there is a specific pulmonary vascular disorder. Indeed, COVID-19 may predispose patients to thrombotic disease in both venous and arterial circulation, and many cases of severe acute pulmonary embolism have been reported. The demonstrated presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) within the endothelial cells suggests that direct viral effects, in addition to indirect effects of perivascular inflammation and coagulopathy, may contribute to pulmonary vasculopathy in COVID-19. In this review, we discuss the pathological mechanisms leading to pulmonary vascular damage during acute infection, which appear to be mainly related to thromboembolic events, an impaired coagulation cascade, micro- and macrovascular thrombosis, endotheliitis and hypoxic pulmonary vasoconstriction. As many patients develop post-COVID symptoms, including dyspnea, we also discuss the hypothesis of pulmonary vascular damage and pulmonary hypertension as a sequela of the infection, which may be involved in the pathophysiology of long COVID.


Sujet(s)
COVID-19 , SARS-CoV-2 , Humains , COVID-19/complications , COVID-19/virologie , COVID-19/anatomopathologie , SARS-CoV-2/pathogénicité , Poumon/vascularisation , Poumon/anatomopathologie , Poumon/virologie , Embolie pulmonaire/virologie , Embolie pulmonaire/étiologie , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/physiopathologie , Hypertension pulmonaire/virologie , Hypertension pulmonaire/anatomopathologie , Syndrome de post-COVID-19 , Thrombose/virologie , Thrombose/étiologie , Thrombose/anatomopathologie
20.
Respir Res ; 25(1): 192, 2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702687

RÉSUMÉ

This review examines how single-cell omics technologies, particularly single-cell RNA sequencing (scRNAseq), enhance our understanding of pulmonary arterial hypertension (PAH). PAH is a multifaceted disorder marked by pulmonary vascular remodeling, leading to high morbidity and mortality. The cellular pathobiology of this heterogeneous disease, involving various vascular and non-vascular cell types, is not fully understood. Traditional PAH studies have struggled to resolve the complexity of pathogenic cell populations. scRNAseq offers a refined perspective by detailing cellular diversity within PAH, identifying unique cell subsets, gene networks, and molecular pathways that drive the disease. We discuss significant findings from recent literature, summarizing how scRNAseq has shifted our understanding of PAH in human, rat, and mouse models. This review highlights the insights gained into cellular phenotypes, gene expression patterns, and novel molecular targets, and contemplates the challenges and prospective paths for research. We propose ways in which single-cell omics could inform future research and translational efforts to combat PAH.


Sujet(s)
Analyse sur cellule unique , Humains , Animaux , Analyse sur cellule unique/méthodes , Hypertension artérielle pulmonaire/génétique , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/physiopathologie , Hypertension artérielle pulmonaire/anatomopathologie , Analyse de séquence d'ARN/méthodes , Hypertension pulmonaire/génétique , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...