Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.905
Filtrer
1.
Clin Sci (Lond) ; 138(13): 817-850, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38920058

RÉSUMÉ

Optimal vascular structure and function are essential for maintaining the physiological functions of the cardiovascular system. Vascular remodelling involves changes in vessel structure, including its size, shape, cellular and molecular composition. These changes result from multiple risk factors and may be compensatory adaptations to sustain blood vessel function. They occur in diverse cardiovascular pathologies, from hypertension to heart failure and atherosclerosis. Dynamic changes in the endothelium, fibroblasts, smooth muscle cells, pericytes or other vascular wall cells underlie remodelling. In addition, immune cells, including macrophages and lymphocytes, may infiltrate vessels and initiate inflammatory signalling. They contribute to a dynamic interplay between cell proliferation, apoptosis, migration, inflammation, and extracellular matrix reorganisation, all critical mechanisms of vascular remodelling. Molecular pathways underlying these processes include growth factors (e.g., vascular endothelial growth factor and platelet-derived growth factor), inflammatory cytokines (e.g., interleukin-1ß and tumour necrosis factor-α), reactive oxygen species, and signalling pathways, such as Rho/ROCK, MAPK, and TGF-ß/Smad, related to nitric oxide and superoxide biology. MicroRNAs and long noncoding RNAs are crucial epigenetic regulators of gene expression in vascular remodelling. We evaluate these pathways for potential therapeutic targeting from a clinical translational perspective. In summary, vascular remodelling, a coordinated modification of vascular structure and function, is crucial in cardiovascular disease pathology.


Sujet(s)
Maladies cardiovasculaires , Hypertension artérielle , Inflammation , Remodelage vasculaire , Humains , Inflammation/métabolisme , Maladies cardiovasculaires/physiopathologie , Maladies cardiovasculaires/métabolisme , Hypertension artérielle/physiopathologie , Hypertension artérielle/métabolisme , Animaux , Stress oxydatif , Transduction du signal , Oxydoréduction
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.
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
4.
Int J Mol Sci ; 25(12)2024 Jun 09.
Article de Anglais | MEDLINE | ID: mdl-38928082

RÉSUMÉ

Vascular remodeling is a very general feature related to angiogenesis and arteriogenesis, which are involved in neovascularization processes [...].


Sujet(s)
Néovascularisation pathologique , Néovascularisation physiologique , Remodelage vasculaire , Humains , Animaux , Néovascularisation pathologique/anatomopathologie , Néovascularisation pathologique/métabolisme ,
5.
Sci Rep ; 14(1): 13287, 2024 06 10.
Article de Anglais | MEDLINE | ID: mdl-38858395

RÉSUMÉ

Clinical outcomes of arteriovenous fistulae (AVF) for hemodialysis remain inadequate since biological mechanisms of AVF maturation and failure are still poorly understood. Aortocaval fistula creation (AVF group) or a sham operation (sham group) was performed in C57BL/6 mice. Venous limbs were collected on postoperative day 7 and total RNA was extracted for high throughput RNA sequencing and bioinformatic analysis. Genes in metabolic pathways were significantly downregulated in the AVF, whereas significant sex differences were not detected. Since gene expression patterns among the AVF group were heterogenous, the AVF group was divided into a 'normal' AVF (nAVF) group and an 'outliers' (OUT) group. The gene expression patterns of the nAVF and OUT groups were consistent with previously published data showing venous adaptive remodeling, whereas enrichment analyses showed significant upregulation of metabolism, inflammation and coagulation in the OUT group compared to the nAVF group, suggesting the heterogeneity during venous remodeling reflects early gene expression changes that may correlate with AVF maturation or failure. Early detection of these processes may be a translational strategy to predict fistula failure and reduce patient morbidity.


Sujet(s)
Anastomose chirurgicale artérioveineuse , Souris de lignée C57BL , Remodelage vasculaire , Animaux , Souris , Mâle , Remodelage vasculaire/génétique , Femelle , Régulation négative/génétique , Veines/métabolisme , Dialyse rénale , Fistule artérioveineuse/génétique , Fistule artérioveineuse/métabolisme , Fistule artérioveineuse/anatomopathologie , Régulation de l'expression des gènes , Analyse de profil d'expression de gènes
6.
Arterioscler Thromb Vasc Biol ; 44(7): 1555-1569, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38779856

RÉSUMÉ

BACKGROUND: ß-aminopropionitrile (BAPN) is a pharmacological inhibitor of LOX (lysyl oxidase) and LOXLs (LOX-like proteins). Administration of BAPN promotes aortopathies, although there is a paucity of data on experimental conditions to generate pathology. The objective of this study was to define experimental parameters and determine whether equivalent or variable aortopathies were generated throughout the aortic tree during BAPN administration in mice. METHODS: BAPN was administered in drinking water for a period ranging from 1 to 12 weeks. The impacts of BAPN were first assessed with regard to BAPN dose, and mouse strain, age, and sex. BAPN-induced aortic pathological characterization was conducted using histology and immunostaining. To investigate the mechanistic basis of regional heterogeneity, the ascending and descending thoracic aortas were harvested after 1 week of BAPN administration before the appearance of overt pathology. RESULTS: BAPN-induced aortic rupture predominantly occurred or originated in the descending thoracic aorta in young C57BL/6J or N mice. No apparent differences were found between male and female mice. For mice surviving 12 weeks of BAPN administration, profound dilatation was consistently observed in the ascending region, while there were more heterogeneous changes in the descending thoracic region. Pathological features were distinct between the ascending and descending thoracic regions. Aortic pathology in the ascending region was characterized by luminal dilatation and elastic fiber disruption throughout the media. The descending thoracic region frequently had dissections with false lumen formation, collagen deposition, and remodeling of the wall surrounding the false lumen. Cells surrounding the false lumen were predominantly positive for α-SMA (α-smooth muscle actin). One week of BAPN administration compromised contractile properties in both regions equivalently, and RNA sequencing did not show obvious differences between the 2 aortic regions in smooth muscle cell markers, cell proliferation markers, and extracellular components. CONCLUSIONS: BAPN-induced pathologies show distinct, heterogeneous features within and between ascending and descending aortic regions in mice.


Sujet(s)
Amino-propionitrile , Aorte thoracique , Rupture aortique , Modèles animaux de maladie humaine , Souris de lignée C57BL , Animaux , Amino-propionitrile/toxicité , Amino-propionitrile/pharmacologie , Aorte thoracique/anatomopathologie , Aorte thoracique/effets des médicaments et des substances chimiques , Aorte thoracique/métabolisme , Femelle , Mâle , Rupture aortique/induit chimiquement , Rupture aortique/anatomopathologie , Rupture aortique/métabolisme , Rupture aortique/prévention et contrôle , Souris , Remodelage vasculaire/effets des médicaments et des substances chimiques , Dilatation pathologique , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/effets des médicaments et des substances chimiques , Muscles lisses vasculaires/métabolisme , Facteurs âges , Facteurs temps , Facteurs sexuels , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lysyloxidase/métabolisme
7.
Arterioscler Thromb Vasc Biol ; 44(7): 1570-1583, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38813697

RÉSUMÉ

BACKGROUND: Pulmonary hypertension (PH) represents an important phenotype in heart failure with preserved ejection fraction (HFpEF). However, management of PH-HFpEF is challenging because mechanisms involved in the regulation of PH-HFpEF remain unclear. METHODS: We used a mass spectrometry-based comparative plasma proteomics approach as a sensitive and comprehensive hypothesis-generating discovery technique to profile proteins in patients with PH-HFpEF and control subjects. We then validated and investigated the role of one of the identified proteins using in vitro cell cultures, in vivo animal models, and independent cohort of human samples. RESULTS: Plasma proteomics identified high protein abundance levels of B2M (ß2-microglobulin) in patients with PH-HFpEF. Interestingly, both circulating and skeletal muscle levels of B2M were increased in mice with skeletal muscle SIRT3 (sirtuin-3) deficiency or high-fat diet-induced PH-HFpEF. Plasma and muscle biopsies from a validation cohort of PH-HFpEF patients were found to have increased B2M levels, which positively correlated with disease severity, especially pulmonary capillary wedge pressure and right atrial pressure at rest. Not only did the administration of exogenous B2M promote migration/proliferation in pulmonary arterial vascular endothelial cells but it also increased PCNA (proliferating cell nuclear antigen) expression and cell proliferation in pulmonary arterial vascular smooth muscle cells. Finally, B2m deletion improved glucose intolerance, reduced pulmonary vascular remodeling, lowered PH, and attenuated RV hypertrophy in mice with high-fat diet-induced PH-HFpEF. CONCLUSIONS: Patients with PH-HFpEF display higher circulating and skeletal muscle expression levels of B2M, the magnitude of which correlates with disease severity. Our findings also reveal a previously unknown pathogenic role of B2M in the regulation of pulmonary vascular proliferative remodeling and PH-HFpEF. These data suggest that circulating and skeletal muscle B2M can be promising targets for the management of PH-HFpEF.


Sujet(s)
Modèles animaux de maladie humaine , Défaillance cardiaque , Hypertension pulmonaire , Protéomique , Débit systolique , bêta-2-Microglobuline , Animaux , Défaillance cardiaque/physiopathologie , Défaillance cardiaque/métabolisme , Défaillance cardiaque/sang , Défaillance cardiaque/génétique , Protéomique/méthodes , Humains , Hypertension pulmonaire/physiopathologie , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/sang , Hypertension pulmonaire/étiologie , Hypertension pulmonaire/génétique , Mâle , Femelle , bêta-2-Microglobuline/génétique , bêta-2-Microglobuline/sang , bêta-2-Microglobuline/métabolisme , Souris de lignée C57BL , Sujet âgé , Sirtuine-3/génétique , Sirtuine-3/métabolisme , Muscles squelettiques/métabolisme , Prolifération cellulaire , Adulte d'âge moyen , Souris , Marqueurs biologiques/sang , Souris knockout , Études cas-témoins , Artère pulmonaire/physiopathologie , Artère pulmonaire/métabolisme , Remodelage vasculaire , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Mouvement cellulaire , Cellules cultivées , Fonction ventriculaire gauche
8.
J Hypertens ; 42(8): 1427-1439, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38690935

RÉSUMÉ

OBJECTIVE: Proliferation and migration of vascular smooth muscle cells (VSMCs) contribute to vascular remodeling. Asprosin, a newly discovered protein hormone, is involved in metabolic diseases. Little is known about the roles of asprosin in cardiovascular diseases. This study focused on the role and mechanism of asprosin on VSMC proliferation and migration, and vascular remodeling in a rat model of hypertension. METHODS AND RESULTS: VSMCs were obtained from the aortic media of 8-week-old male Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Asprosin was upregulated in the VSMCs of SHR. For in vitro studies, asprosin promoted VSMC proliferation and migration of WKY and SHR, and increased Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity, NOX1/2/4 protein expressions and superoxide production. Knockdown of asprosin inhibited the proliferation, migration, NOX activity, NOX1/2 expressions and superoxide production in the VSMCs of SHR. The roles of asprosin in promoting VSMC proliferation and migration were not affected by hydrogen peroxide scavenger, but attenuated by superoxide scavenger, selective NOX1 or NOX2 inhibitor. Toll-like receptor 4 (TLR4) was upregulated in SHR, TLR4 knockdown inhibited asprosin overexpression-induced proliferation, migration and oxidative stress in VSMCs of WKY and SHR. Asprosin was upregulated in arteries of SHR, and knockdown of asprosin in vivo not only attenuated oxidative stress and vascular remodeling in aorta and mesentery artery, but also caused a subsequent persistent antihypertensive effect in SHR. CONCLUSIONS: Asprosin promotes VSMC proliferation and migration via NOX-mediated superoxide production. Inhibition of endogenous asprosin expression attenuates VSMC proliferation and migration, and vascular remodeling of SHR.


Sujet(s)
Mouvement cellulaire , Prolifération cellulaire , Hypertension artérielle , Muscles lisses vasculaires , Rats de lignée SHR , Rats de lignée WKY , Transduction du signal , Superoxydes , Remodelage vasculaire , Animaux , Mâle , Superoxydes/métabolisme , Rats , Hypertension artérielle/métabolisme , Hypertension artérielle/physiopathologie , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Myocytes du muscle lisse/métabolisme , NADPH oxidase/métabolisme , Hormones peptidiques/métabolisme , Fibrilline-1/métabolisme , Récepteur de type Toll-4/métabolisme
9.
Curr Top Dev Biol ; 159: 344-370, 2024.
Article de Anglais | MEDLINE | ID: mdl-38729681

RÉSUMÉ

The development of the vascular system is crucial in supporting the growth and health of all other organs in the body, and vascular system dysfunction is the major cause of human morbidity and mortality. This chapter discusses three successive processes that govern vascular system development, starting with the differentiation of the primitive vascular system in early embryonic development, followed by its remodeling into a functional circulatory system composed of arteries and veins, and its final maturation and acquisition of an organ specific semi-permeable barrier that controls nutrient uptake into tissues and hence controls organ physiology. Along these steps, endothelial cells forming the inner lining of all blood vessels acquire extensive heterogeneity in terms of gene expression patterns and function, that we are only beginning to understand. These advances contribute to overall knowledge of vascular biology and are predicted to unlock the unprecedented therapeutic potential of the endothelium as an avenue for treatment of diseases associated with dysfunctional vasculature.


Sujet(s)
Remodelage vasculaire , Humains , Animaux , Vaisseaux sanguins/croissance et développement , Vaisseaux sanguins/métabolisme , Vaisseaux sanguins/embryologie , Néovascularisation physiologique , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Cellules endothéliales/physiologie , Différenciation cellulaire , Développement embryonnaire , Endothélium vasculaire/cytologie
10.
Circ Res ; 135(1): 76-92, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38747146

RÉSUMÉ

BACKGROUND: Hypoxia and oxidative stress contribute to the development of pulmonary hypertension (PH). tRNA-derived fragments play important roles in RNA interference and cell proliferation, but their epitranscriptional roles in PH development have not been investigated. We aimed to gain insight into the mechanistic contribution of oxidative stress-induced 8-oxoguanine in pulmonary vascular remodeling. METHODS: Through small RNA modification array analysis and quantitative polymerase chain reaction, a significant upregulation of the 8-oxoguanine -modified tRF-1-AspGTC was found in the lung tissues and the serum of patients with PH. RESULTS: This modification occurs at the position 5 of the tRF-1-AspGTC (5o8G tRF). Inhibition of the 5o8G tRF reversed hypoxia-induced proliferation and apoptosis resistance in pulmonary artery smooth muscle cells. Further investigation unveiled that the 5o8G tRF retargeted mRNA of WNT5A (Wingless-type MMTV integration site family, member 5A) and CASP3 (Caspase3) and inhibited their expression. Ultimately, BMPR2 (Bone morphogenetic protein receptor 2) -reactive oxygen species/5o8G tRF/WNT5A signaling pathway exacerbated the progression of PH. CONCLUSIONS: Our study highlights the role of site-specific 8-oxoguanine-modified tRF in promoting the development of PH. Our findings present a promising therapeutic avenue for managing PH and propose 5o8G tRF as a potential innovative marker for diagnosing this disease.


Sujet(s)
Marqueurs biologiques , Récepteurs de la protéine morphogénique osseuse de type II , Hypertension pulmonaire , Artère pulmonaire , Hypertension pulmonaire/métabolisme , Hypertension pulmonaire/génétique , Hypertension pulmonaire/étiologie , Humains , Récepteurs de la protéine morphogénique osseuse de type II/métabolisme , Récepteurs de la protéine morphogénique osseuse de type II/génétique , Animaux , Marqueurs biologiques/métabolisme , Marqueurs biologiques/sang , Artère pulmonaire/métabolisme , Protéine Wnt-5a/métabolisme , Protéine Wnt-5a/génétique , Guanine/analogues et dérivés , Guanine/métabolisme , Mâle , Stress oxydatif , Caspase-3/métabolisme , Myocytes du muscle lisse/métabolisme , Prolifération cellulaire , Apoptose , Cellules cultivées , Remodelage vasculaire , Femelle , Rats , Espèces réactives de l'oxygène/métabolisme , Muscles lisses vasculaires/métabolisme
11.
Vascul Pharmacol ; 155: 107379, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38762131

RÉSUMÉ

Pulmonary hypertension (PH) is a progressive, severe and to date not curable disease of the pulmonary vasculature. Alterations of the insulin-like growth factor 1 (IGF-1) system are known to play a role in vascular pathologies and IGF-binding proteins (IGFBPs) are important regulators of the bioavailability and function of IGFs. In this study, we show that circulating plasma levels of IGFBP-1, IGFBP-2 and IGFBP-3 are increased in idiopathic pulmonary arterial hypertension (IPAH) patients compared to healthy individuals. These binding proteins inhibit the IGF-1 induced IGF-1 receptor (IGF1R) phosphorylation and exhibit diverging effects on the IGF-1 induced signaling pathways in human pulmonary arterial cells (i.e. healthy as well as IPAH-hPASMCs, and healthy hPAECs). Furthermore, IGFBPs are differentially expressed in an experimental mouse model of PH. In hypoxic mouse lungs, IGFBP-1 mRNA expression is decreased whereas the mRNA for IGFBP-2 is increased. In contrast to IGFBP-1, IGFBP-2 shows vaso-constrictive properties in the murine pulmonary vasculature. Our analyses show that IGFBP-1 and IGFBP-2 exhibit diverging effects on IGF-1 signaling and display a unique IGF1R-independent kinase activation pattern in human pulmonary arterial smooth muscle cells (hPASMCs), which represent a major contributor of PAH pathobiology. Furthermore, we could show that IGFBP-2, in contrast to IGFBP-1, induces epidermal growth factor receptor (EGFR) signaling, Stat-3 activation and expression of Stat-3 target genes. Based on our results, we conclude that the IGFBP family, especially IGFBP-1, IGFBP-2 and IGFBP-3, are deregulated in PAH, that they affect IGF signaling and thereby regulate the cellular phenotype in PH.


Sujet(s)
Modèles animaux de maladie humaine , Protéine-1 de liaison aux IGF , Protéine-2 de liaison aux IGF , Protéine-3 de liaison aux IGF , Facteur de croissance IGF-I , Myocytes du muscle lisse , Artère pulmonaire , Récepteur IGF de type 1 , Transduction du signal , Humains , Animaux , Récepteur IGF de type 1/métabolisme , Récepteur IGF de type 1/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Artère pulmonaire/physiopathologie , Protéine-3 de liaison aux IGF/métabolisme , Protéine-3 de liaison aux IGF/génétique , Protéine-2 de liaison aux IGF/métabolisme , Protéine-2 de liaison aux IGF/génétique , Facteur de croissance IGF-I/métabolisme , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/anatomopathologie , Cellules cultivées , Mâle , Protéine-1 de liaison aux IGF/métabolisme , Protéine-1 de liaison aux IGF/génétique , Phosphorylation , Facteur de transcription STAT-3/métabolisme , Études cas-témoins , Souris de lignée C57BL , Hypertension artérielle pulmonaire primitive familiale/métabolisme , Hypertension artérielle pulmonaire primitive familiale/physiopathologie , Hypertension artérielle pulmonaire primitive familiale/anatomopathologie , Hypertension artérielle pulmonaire primitive familiale/génétique , Femelle , Récepteurs ErbB/métabolisme , Adulte d'âge moyen , Remodelage vasculaire , Adulte , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie
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.
Hypertension ; 81(7): 1524-1536, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38716674

RÉSUMÉ

BACKGROUND: Preeclampsia is a significant pregnancy disorder with an unknown cause, mainly attributed to impaired spiral arterial remodeling. METHODS: Using RNA sequencing, we identified key genes in placental tissues from healthy individuals and preeclampsia patients. Placenta and plasma samples from pregnant women were collected to detect the expression of TPBG (trophoblast glycoprotein). Pregnant rats were injected with TPBG-carrying adenovirus to detect preeclamptic features. HTR-8/SVneo cells transfected with a TPBG overexpression lentiviral vector were used in cell function experiments. The downstream molecular mechanisms of TPBG were explored using RNA sequencing and single-cell RNA sequencing data. TPBG expression was knocked down in the lipopolysaccharide-induced preeclampsia-like rat model to rescue the preeclampsia features. We also assessed TPBG's potential as an early preeclampsia predictor using clinical plasma samples. RESULTS: TPBG emerged as a crucial differentially expressed gene, expressed specifically in syncytiotrophoblasts and extravillous trophoblasts. Subsequently, we established a rat model with preeclampsia-like phenotypes by intravenously injecting TPBG-expressing adenoviruses, observing impaired spiral arterial remodeling, thus indicating a causal correlation between TPBG overexpression and preeclampsia. Studies with HTR-8/SVneo cells, chorionic villous explants, and transwell assays showed TPBG overexpression disrupts trophoblast/extravillous trophoblast migration/invasion and chemotaxis. Notably, TPBG knockdown alleviated the lipopolysaccharide-induced preeclampsia-like rat model. We enhanced preeclampsia risk prediction in early gestation by combining TPBG expression with established clinical predictors. CONCLUSIONS: These findings are the first to show that TPBG overexpression contributes to preeclampsia development by affecting uterine spiral artery remodeling. We propose TPBG levels in maternal blood as a predictor of preeclampsia risk. The proposed mechanism by which TPBG overexpression contributes to the occurrence of preeclampsia via its disruptive effect on trophoblast and extravillous trophoblast migration/invasion on uterine spiral artery remodeling, thereby increasing the risk of preeclampsia.


Sujet(s)
Mouvement cellulaire , Pré-éclampsie , Trophoblastes , Femelle , Pré-éclampsie/génétique , Pré-éclampsie/métabolisme , Grossesse , Trophoblastes/métabolisme , Animaux , Rats , Humains , Modèles animaux de maladie humaine , Artère utérine/métabolisme , Artère utérine/anatomopathologie , Rat Sprague-Dawley , Remodelage vasculaire/physiologie , Remodelage vasculaire/génétique , Placenta/métabolisme , Glycoprotéines/génétique , Glycoprotéines/métabolisme , Adulte
16.
Sci Rep ; 14(1): 12431, 2024 05 30.
Article de Anglais | MEDLINE | ID: mdl-38816406

RÉSUMÉ

Pulmonary arterial hypertension (PAH) is a fatal disease featured by high morbidity and mortality. Although Cordycepin is known for its anti-inflammatory, antioxidant and immune-enhancing effects, its role in PAH treatment and the underlying mechanisms remain unclear. The therapeutic effects of Cordycepin on rats with PAH were investigated using a monocrotaline (MCT)-induced rat model. The metabolic effects of Cordycepin were assessed based on the plasma metabolome. The potential mechanisms of Cordycepin in PAH treatment were investigated through transcriptome sequencing and validated in pulmonary artery smooth muscle cells (PASMC). Evaluations included hematoxylin and eosin staining for pulmonary vascular remodeling, CCK-8 assay, EDU, and TUNEL kits for cell viability, proliferation, and apoptosis, respectively, and western blot for protein expression. Cordycepin significantly reduced right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) in PAH rats, and mitigated pulmonary vascular remodeling. Plasma metabolomics showed that Cordycepin could reverse the metabolic disorders in the lungs of MCT-induced PAH rats, particularly impacting linoleic acid and alpha-linolenic acid metabolism pathways. Transcriptomics revealed that the P53 pathway might be the primary pathway involved, and western blot results showed that Cordycepin significantly increased P53 and P21 protein levels in lung tissues. Integrated analysis of transcriptomics and metabolomics suggested that these pathways were mainly enriched in linoleic acid metabolism and alpha-linolenic acid metabolism pathway. In vitro experiments demonstrated that Cordycepin significantly inhibited the PDGFBB (PD)-induced abnormal proliferation and migration of PASMC and promoted PD-induced apoptosis. Meanwhile, Cordycepin enhanced the expression levels of P53 and P21 proteins in PD-insulted PASMC. However, inhibitors of P53 and P21 eliminated these effects of Cordycepin. Cordycepin may activate the P53-P21 pathway to inhibit abnormal proliferation and migration of PASMC and promote apoptosis, offering a potential approach for PAH treatment.


Sujet(s)
Apoptose , Prolifération cellulaire , Désoxyadénosine , Hypertension artérielle pulmonaire , Animaux , Désoxyadénosine/pharmacologie , Désoxyadénosine/usage thérapeutique , Rats , Mâle , Apoptose/effets des médicaments et des substances chimiques , Hypertension artérielle pulmonaire/traitement médicamenteux , Hypertension artérielle pulmonaire/métabolisme , Hypertension artérielle pulmonaire/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transcriptome/effets des médicaments et des substances chimiques , Métabolomique , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques , Monocrotaline , Artère pulmonaire/effets des médicaments et des substances chimiques , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Remodelage vasculaire/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Acide linoléique/pharmacologie , Hypertrophie ventriculaire droite/traitement médicamenteux , Hypertrophie ventriculaire droite/métabolisme , Analyse de profil d'expression de gènes
17.
Basic Res Cardiol ; 119(3): 371-395, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38700707

RÉSUMÉ

Ascending thoracic aortic aneurysm (ATAA) remains a significant medical concern, with its asymptomatic nature posing diagnostic and monitoring challenges, thereby increasing the risk of aortic wall dissection and rupture. Current management of aortic repair relies on an aortic diameter threshold. However, this approach underestimates the complexity of aortic wall disease due to important knowledge gaps in understanding its underlying pathologic mechanisms.Since traditional risk factors cannot explain the initiation and progression of ATAA leading to dissection, local vascular factors such as extracellular matrix (ECM) and vascular smooth muscle cells (VSMCs) might harbor targets for early diagnosis and intervention. Derived from diverse embryonic lineages, VSMCs exhibit varied responses to genetic abnormalities that regulate their contractility. The transition of VSMCs into different phenotypes is an adaptive response to stress stimuli such as hemodynamic changes resulting from cardiovascular disease, aging, lifestyle, and genetic predisposition. Upon longer exposure to stress stimuli, VSMC phenotypic switching can instigate pathologic remodeling that contributes to the pathogenesis of ATAA.This review aims to illuminate the current understanding of cellular and molecular characteristics associated with ATAA and dissection, emphasizing the need for a more nuanced comprehension of the impaired ECM-VSMC network.


Sujet(s)
Anévrysme de l'aorte thoracique , , Muscles lisses vasculaires , Myocytes du muscle lisse , Humains , Anévrysme de l'aorte thoracique/anatomopathologie , Anévrysme de l'aorte thoracique/génétique , Anévrysme de l'aorte thoracique/métabolisme , Anévrysme de l'aorte thoracique/physiopathologie , /anatomopathologie , /génétique , /métabolisme , Animaux , Muscles lisses vasculaires/anatomopathologie , Muscles lisses vasculaires/métabolisme , Myocytes du muscle lisse/anatomopathologie , Myocytes du muscle lisse/métabolisme , Aorte thoracique/anatomopathologie , Aorte thoracique/physiopathologie , Remodelage vasculaire , Matrice extracellulaire/anatomopathologie , Matrice extracellulaire/métabolisme , Phénotype
18.
Cell Mol Biol Lett ; 29(1): 69, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38741032

RÉSUMÉ

BACKGROUND: Pulmonary hypertension (PH) is a progressive disease characterized by pulmonary vascular remodeling. Increasing evidence indicates that endothelial-to-mesenchymal transition (EndMT) in pulmonary artery endothelial cells (PAECs) is a pivotal trigger initiating this remodeling. However, the regulatory mechanisms underlying EndMT in PH are still not fully understood. METHODS: Cytokine-induced hPAECs were assessed using RNA methylation quantification, qRT-PCR, and western blotting to determine the involvement of N6-methyladenosine (m6A) methylation in EndMT. Lentivirus-mediated silencing, overexpression, tube formation, and wound healing assays were utilized to investigate the function of METTL3 in EndMT. Endothelial-specific gene knockout, hemodynamic measurement, and immunostaining were performed to explore the roles of METTL3 in pulmonary vascular remodeling and PH. RNA-seq, RNA Immunoprecipitation-based qPCR, mRNA stability assay, m6A mutation, and dual-luciferase assays were employed to elucidate the mechanisms of RNA methylation in EndMT. RESULTS: The global levels of m6A and METTL3 expression were found to decrease in TNF-α- and TGF-ß1-induced EndMT in human PAECs (hPAECs). METTL3 inhibition led to reduced endothelial markers (CD31 and VE-cadherin) and increased mesenchymal markers (SM22 and N-cadherin) as well as EndMT-related transcription factors (Snail, Zeb1, Zeb2, and Slug). The endothelial-specific knockout of Mettl3 promoted EndMT and exacerbated pulmonary vascular remodeling and hypoxia-induced PH (HPH) in mice. Mechanistically, METTL3-mediated m6A modification of kruppel-like factor 2 (KLF2) plays a crucial role in the EndMT process. KLF2 overexpression increased CD31 and VE-cadherin levels while decreasing SM22, N-cadherin, and EndMT-related transcription factors, thereby mitigating EndMT in PH. Mutations in the m6A site of KLF2 mRNA compromise KLF2 expression, subsequently diminishing its protective effect against EndMT. Furthermore, KLF2 modulates SM22 expression through direct binding to its promoter. CONCLUSIONS: Our findings unveil a novel METTL3/KLF2 pathway critical for protecting hPAECs against EndMT, highlighting a promising avenue for therapeutic investigation in PH.


Sujet(s)
Adénosine , Cellules endothéliales , Transition épithélio-mésenchymateuse , Hypertension pulmonaire , Facteurs de transcription Krüppel-like , Methyltransferases , Adénosine/analogues et dérivés , Adénosine/métabolisme , Animaux , Hypertension pulmonaire/génétique , Hypertension pulmonaire/métabolisme , Humains , Methyltransferases/métabolisme , Methyltransferases/génétique , Souris , Cellules endothéliales/métabolisme , Transition épithélio-mésenchymateuse/génétique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Artère pulmonaire/métabolisme , Artère pulmonaire/anatomopathologie , Méthylation , Souris de lignée C57BL , Cadhérines/métabolisme , Cadhérines/génétique , Mâle , Remodelage vasculaire/génétique , Cellules cultivées
19.
Genome Biol ; 25(1): 117, 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38715110

RÉSUMÉ

BACKGROUND: Preeclampsia, one of the most lethal pregnancy-related diseases, is associated with the disruption of uterine spiral artery remodeling during placentation. However, the early molecular events leading to preeclampsia remain unknown. RESULTS: By analyzing placentas from preeclampsia, non-preeclampsia, and twin pregnancies with selective intrauterine growth restriction, we show that the pathogenesis of preeclampsia is attributed to immature trophoblast and maldeveloped endothelial cells. Delayed epigenetic reprogramming during early extraembryonic tissue development leads to generation of excessive immature trophoblast cells. We find reduction of de novo DNA methylation in these trophoblast cells results in selective overexpression of maternally imprinted genes, including the endoretrovirus-derived gene PEG10 (paternally expressed gene 10). PEG10 forms virus-like particles, which are transferred from the trophoblast to the closely proximate endothelial cells. In normal pregnancy, only a low amount of PEG10 is transferred to maternal cells; however, in preeclampsia, excessive PEG10 disrupts maternal vascular development by inhibiting TGF-beta signaling. CONCLUSIONS: Our study reveals the intricate epigenetic mechanisms that regulate trans-generational genetic conflict and ultimately ensure proper maternal-fetal interface formation.


Sujet(s)
Pré-éclampsie , Trophoblastes , Remodelage vasculaire , Pré-éclampsie/génétique , Grossesse , Femelle , Humains , Trophoblastes/métabolisme , Remodelage vasculaire/génétique , Placenta/métabolisme , Méthylation de l'ADN , Épigenèse génétique , Cellules endothéliales/métabolisme , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Empreinte génomique , Facteur de croissance transformant bêta/métabolisme , Retard de croissance intra-utérin/génétique , Placentation/génétique , Protéines de liaison à l'ARN , Protéines régulatrices de l'apoptose
20.
FASEB J ; 38(9): e23645, 2024 May 15.
Article de Anglais | MEDLINE | ID: mdl-38703043

RÉSUMÉ

Inflammation assumes a pivotal role in the aortic remodeling of aortic dissection (AD). Asiatic acid (AA), a triterpene compound, is recognized for its strong anti-inflammatory properties. Yet, its effects on ß-aminopropionitrile (BAPN)-triggered AD have not been clearly established. The objective is to determine whether AA attenuates adverse aortic remodeling in BAPN-induced AD and clarify potential molecular mechanisms. In vitro studies, RAW264.7 cells pretreated with AA were challenged with lipopolysaccharide (LPS), and then the vascular smooth muscle cells (VSMCs)-macrophage coculture system was established to explore intercellular interactions. To induce AD, male C57BL/6J mice at three weeks of age were administered BAPN at a dosage of 1 g/kg/d for four weeks. To decipher the mechanism underlying the effects of AA, RNA sequencing analysis was conducted, with subsequent validation of these pathways through cellular experiments. AA exhibited significant suppression of M1 macrophage polarization. In the cell coculture system, AA facilitated the transformation of VSMCs into a contractile phenotype. In the mouse model of AD, AA strikingly prevented the BAPN-induced increases in inflammation cell infiltration and extracellular matrix degradation. Mechanistically, RNA sequencing analysis revealed a substantial upregulation of CX3CL1 expression in BAPN group but downregulation in AA-treated group. Additionally, it was observed that the upregulation of CX3CL1 negated the beneficial impact of AA on the polarization of macrophages and the phenotypic transformation of VSMCs. Crucially, our findings revealed that AA is capable of downregulating CX3CL1 expression, accomplishing this by obstructing the nuclear translocation of NF-κB p65. The findings indicate that AA holds promise as a prospective treatment for adverse aortic remodeling by suppressing the activity of NF-κB p65/CX3CL1 signaling pathway.


Sujet(s)
, Chimiokine CX3CL1 , Souris de lignée C57BL , Triterpènes pentacycliques , Transduction du signal , Facteur de transcription RelA , Remodelage vasculaire , Animaux , Souris , Mâle , /métabolisme , /anatomopathologie , /traitement médicamenteux , Triterpènes pentacycliques/pharmacologie , Remodelage vasculaire/effets des médicaments et des substances chimiques , Cellules RAW 264.7 , Transduction du signal/effets des médicaments et des substances chimiques , Facteur de transcription RelA/métabolisme , Chimiokine CX3CL1/métabolisme , Chimiokine CX3CL1/génétique , Muscles lisses vasculaires/métabolisme , Muscles lisses vasculaires/anatomopathologie , Amino-propionitrile/pharmacologie , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Myocytes du muscle lisse/métabolisme , Myocytes du muscle lisse/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...