human pulmonary artery
Recently Published Documents


TOTAL DOCUMENTS

372
(FIVE YEARS 63)

H-INDEX

41
(FIVE YEARS 4)

2021 ◽  
Vol 42 (Supplement_1) ◽  
Author(s):  
R Ramadhiani ◽  
K Ikeda ◽  
K Miyagawa ◽  
G R T Ryanto ◽  
N Tamada ◽  
...  

Abstract Background Despite recently developed clinical therapies, vascular remodelling in pulmonary arterial hypertension (PAH) progressively worsen. Hemodynamic unloading has been proposed to normalize the remodelled pulmonary vascular structures in the lungs. Recently, it has been reported that cellular senescence was associated with the irreversibility of pulmonary vascular structures after hemodynamic unloading. Purpose This study aims to elucidate the role of senescent endothelial cells (ECs) in the pathogenesis of PAH. Methods We generated EC-specific progeroid mice in which ECs undergo premature senescence by overexpressing the dominant-negative form of telomere repeat-binding factor 2 under the control of the VE-cadherin promoter. Following three weeks of hypoxia exposure, the PH phenotypes were assessed by RVSP, lung histology, and RT-qPCR. The interaction of human pulmonary artery ECs (hPAECs) and human pulmonary artery smooth muscle cells (hPASMCs) was indirectly and directly explored through the co-culture system. Gamma-secretase inhibitor (DAPT) was administrated to inhibit Notch signalling both in the in-vitro and in-vivo study. Results EC-specific progeroid mice showed exacerbated pulmonary hypertension after chronic hypoxia exposure, accompanied by the enhanced medial SMCs proliferation in the distal pulmonary arteries. Contact-mediated interaction with senescent hPAECs increased proliferation and migration capacities in hPASMCs, while no such effects were detected in the absence of ECs-SMCs contact. Consistently, senescent ECs highly expressed Notch ligands, thus activated Notch signalling in hPASMCs, leading to increased Notch target genes in hPASMCs. Pharmacological inhibition of Notch signalling attenuated the enhanced SMCs proliferation and migration induced by senescent hPAECs, as well as the worsened PH phenotypes in EC-specific progeroid mice. Conclusions Our data established a crucial role of senescent ECs in the PAH pathogenesis through the dysregulated SMC functions via juxtacrine signaling. Senescent ECs are attracting targets for further pathological-targeted therapy to cure PAH completely. FUNDunding Acknowledgement Type of funding sources: None.


Author(s):  
Shouxian Wang ◽  
Yu Wang ◽  
Chang Liu ◽  
Gang Xu ◽  
Wenxiang Gao ◽  
...  

EPAS1 (endothelial PAS domain protein 1), as the major effect gene for the adaptation to chronic hypoxia, is required for hypoxic pulmonary hypertension (HPH). Downregulated EPAS1 ameliorates the development of HPH. We confirmed that EPAS1-specific inhibitor PT2385 ameliorated HPH features, as demonstrated by right ventricle hypertrophy, right ventricular systolic pressure, and pulmonary vascular remodeling. However, the mechanism of EPAS1 in HPH pathogenesis remains unclear. RNA sequencing in human pulmonary artery endothelial cells with EPAS1 knockdown identified EPAS1-regulated genes, including ICAM1 (intercellular adhesion molecule 1), which created a proinflammatory perivascular microenvironment associated with HPH by elevating leukocyte adhesion to the vascular endothelium. Chromatin immunoprecipitation assays revealed that EPAS1 directly bound to ICAM1 promoter. The long noncoding RNA small nucleolar RNA host gene 5 (SNHG5), significantly increased in acute exacerbation period of chronic obstructive pulmonary disease and hypoxic human pulmonary artery endothelial cells, also contributed to the regulation of ICAM1 expression. Endothelial-specific deletion of Snhg5 also rescued HPH in mice. Overexpression of EPAS1 or SNHG5 enhanced, while the depletion of EPAS1 or SNHG5 attenuated, ICAM1 transactivation. SNHG5 was directly regulated by EPAS1, and interestingly, the upregulated SNHG5 could further enhance the levels of EPAS1, which consequently led to hypoxia-induced ICAM1 transactivation. RNA pull-down assay followed by high-throughput sequencing demonstrated that miR-625-5p could bind to SNHG5. Manipulating miR-625-5p altered the levels of EPAS1 during hypoxia. Our data showed a positive feed-forward exists between EPAS1 and SNHG5 signaling during hypoxia-induced ICAM1 transactivation in endothelial cells. Targeting EPAS1 and SNHG5 may provide promising strategies for the prevention of HPH.


2021 ◽  
Vol 2021 ◽  
pp. 1-15
Author(s):  
Cuilan Hou ◽  
Juan Chen ◽  
Yuqi Zhao ◽  
Yanhua Niu ◽  
Shujia Lin ◽  
...  

Aims. This study is aimed at examining whether fatty acid synthase (FAS) can regulate mitochondrial function in hypoxia-induced pulmonary arterial hypertension (PAH) and its related mechanism. Results. The expression of FAS significantly increased in the lung tissue of mice with hypoxia-induced PAH, and its pharmacological inhibition by C75 ameliorated right ventricle cardiac function as revealed by echocardiographic analysis. Based on transmission electron microscopy and Seahorse assays, the mitochondrial function of mice with hypoxia was abnormal but was partially reversed after C75 injection. In vitro studies also showed an increase in the expression of FAS in hypoxia-induced human pulmonary artery smooth muscle cells (HPASMCs), which could be attenuated by FAS shRNA as well as C75 treatment. Meanwhile, C75 treatment reversed hypoxia-induced oxidative stress and activated PI3K/AKT signaling. shRNA-mediated inhibition of FAS reduced its expression and oxidative stress levels and improved mitochondrial respiratory capacity and ATP levels of hypoxia-induced HPASMCs. Conclusions. Inhibition of FAS plays a crucial role in shielding mice from hypoxia-induced PAH, which was partially achieved through the activation of PI3K/AKT signaling, indicating that the inhibition of FAS may provide a potential future direction for reversing PAH in humans.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Li Yi ◽  
JunFang Liu ◽  
Ming Deng ◽  
Huihua Zuo ◽  
Mingyan Li

Abstract Objective This study aimed to determine the effects of emodin on the viability, proliferation and apoptosis of human pulmonary artery smooth muscle cells (PASMCs) under hypoxia and to explore the underling molecular mechanisms. Methods PASMCs were cultured in a hypoxic environment (1% oxygen) and then treated with emodin. Cell viability, proliferation and apoptosis were evaluated using CCK-8 assay, EdU staining assay, western blot and Mito-tracker red CMXRos and Annexin V-FITC apoptosis detection assay. The microRNA (miRNA)/mRNA and protein expression levels were assessed by quantitative real-time PCR and western blotting, respectively. Based on transcriptomics and proteomics were used to identify potential signaling pathways. Luciferase reporter assay was utilized to examine the interaction between miR-244-5p and DEGS1. Results Emodin at 40 and 160 µM concentration-dependently suppressed cell viability, proliferation and migration, but enhanced cell apoptosis of PASMCs under hypoxia. Transcriptomic and proteomic analysis revealed that emodin could attenuate the activity of PI3K/Akt signaling in PASMCs under hypoxia. In addition, delta 4-desaturase, sphingolipid 1 (DEGS1) was found to be a direct target of miR-244-5p. Emodin could significantly up-regulated miR-244-5p expression and down-regulated DEGS1 expression in PASMCs under hypoxia. Furthermore, emodin-mediated effects on cell viability, migration, apoptosis and PI3K/Akt signaling activity of PASMCs under hypoxia were significantly attenuated by miR-244-5p knockdown. Conclusions Our results indicated that emodin suppressed cell viability, proliferation and migration, promoted cell apoptosis of PASMCs under hypoxia via modulating miR-244-5p-mediated DEGS1/PI3K/Akt signaling pathway. MiR-244-5p/DEGS1 axis was initially investigated in this current study, which is expected to further the understanding of the etiology of pulmonary arterial hypertension.


Author(s):  
Sanghamitra Sahoo ◽  
Yao Li ◽  
Daniel de Jesus ◽  
John Charles Sembrat II ◽  
Mauricio M Rojas ◽  
...  

Pulmonary arterial hypertension (PAH) is a fatal cardiopulmonary disease characterized by increased vascular cell proliferation with resistance to apoptosis and occlusive remodeling of the small pulmonary arteries in humans. The Notch family of proteins are proximal signaling mediators of an evolutionarily conserved pathway that effect cell proliferation, fate determination, and development. In endothelial cells (ECs), Notch receptor 2 (Notch2) has been shown to promote endothelial apoptosis. However, a pro- or anti-proliferative role for Notch2 in pulmonary endothelial proliferation and ensuing PAH is unknown. Herein, we postulated that suppressed Notch2 signaling drives pulmonary endothelial proliferation in the setting of PAH. We observed that levels of Notch2 are ablated in lung and PA tissue samples from PAH patients compared to non-PAH controls. Interestingly, Notch2 expression was attenuated in human pulmonary artery endothelial cells (hPAECs) exposed to vasoactive factors including hypoxia, TGFβ, ET-1, and IGF-1. Gene silencing of Notch2 increased EC proliferation and reduced apoptosis. At the molecular level, Notch2-deficient hPAECs activated Akt, Erk1/2 and anti-apoptotic protein Bcl-2, and reduced levels of p21cip and Bax. Intriguingly, loss of Notch2 elicits a paradoxical activation of Notch1 and transcriptional upregulation of canonical Notch target genes Hes1, Hey1 and Hey2. Further, reduction in Rb and increased E2F1 binding to the Notch1 promoter appear to explain the upregulation of Notch1. In aggregate, our results demonstrate that loss of Notch2 derepresses Notch1 and elicits aberrant EC hallmarks of PAH. The data underscore a novel role for Notch in the maintenance of endothelial cell homeostasis.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Marta Targosz-Korecka ◽  
Agata Kubisiak ◽  
Damian Kloska ◽  
Aleksandra Kopacz ◽  
Anna Grochot-Przeczek ◽  
...  

AbstractEndothelial cells (ECs) play a crucial role in the development and propagation of the severe COVID-19 stage as well as multiorgan dysfunction. It remains, however, controversial whether COVID-19-induced endothelial injury is caused directly by the infection of ECs with SARS-CoV-2 or via indirect mechanisms. One of the major concerns is raised by the contradictory data supporting or denying the presence of ACE2, the SARS-CoV-2 binding receptor, on the EC surface. Here, we show that primary human pulmonary artery ECs possess ACE2 capable of interaction with the viral Spike protein (S-protein) and demonstrate the crucial role of the endothelial glycocalyx in the regulation of the S-protein binding to ACE2 on ECs. Using force spectroscopy method, we directly measured ACE2- and glycocalyx-dependent adhesive forces between S-protein and ECs and characterized the nanomechanical parameters of the cells exposed to S-protein. We revealed that the intact glycocalyx strongly binds S-protein but screens its interaction with ACE2. Reduction of glycocalyx layer exposes ACE2 receptors and promotes their interaction with S-protein. These results indicate that the susceptibility of ECs to COVID-19 infection may depend on the glycocalyx condition.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yi Na Dong ◽  
Fu-Chun Hsu ◽  
Cynthia J. Koziol-White ◽  
Victoria Stepanova ◽  
Joseph Jude ◽  
...  

AbstractN-methyl-d-aspartate (NMDA) receptors are widely expressed in the central nervous system. However, their presence and function at extraneuronal sites is less well characterized. In the present study, we examined the expression of NMDA receptor subunit mRNA and protein in human pulmonary artery (HPA) by quantitative polymerase chain reaction (PCR), immunohistochemistry and immunoblotting. We demonstrate that both GluN1 and GluN2 subunit mRNAs are expressed in HPA. In addition, GluN1 and GluN2 (A–D) subunit proteins are expressed by human pulmonary artery smooth muscle cells (HPASMCs) in vitro and in vivo. These subunits localize on the surface of HPASMCs and form functional ion channels as evidenced by whole-cell patch-clamp electrophysiology and reduced phenylephrine-induced contractile responsiveness of human pulmonary artery by the NMDA receptor antagonist MK801 under hypoxic condition. HPASMCs also express high levels of serine racemase and vesicular glutamate transporter 1, suggesting a potential source of endogenous agonists for NMDA receptor activation. Our findings show HPASMCs express functional NMDA receptors in line with their effect on pulmonary vasoconstriction, and thereby suggest a novel therapeutic target for pharmacological modulations in settings associated with pulmonary vascular dysfunction.


Sign in / Sign up

Export Citation Format

Share Document