Treatment with Inhaled IL10 Decreases Pulmonary Arterial Hypertension in Slit3 Knockout Mice with Congenital Diaphragmatic Hernia

2016 ◽  
Vol 223 (4) ◽  
pp. S92
Author(s):  
Mansi Shah ◽  
Michael R. Phillips ◽  
Swathi Balaji ◽  
Sundeep G. Keswani ◽  
Sean E. McLean
2015 ◽  
Vol 45 (5) ◽  
pp. 572-577 ◽  
Author(s):  
E. Spaggiari ◽  
J. J. Stirnemann ◽  
P. Sonigo ◽  
N. Khen-Dunlop ◽  
L. De Saint Blanquat ◽  
...  

2008 ◽  
Vol 21 (3) ◽  
pp. 507-515 ◽  
Author(s):  
Carsten Wunderlich ◽  
Alexander Schmeisser ◽  
Christian Heerwagen ◽  
Bernd Ebner ◽  
Kristin Schober ◽  
...  

2019 ◽  
Vol 125 (10) ◽  
pp. 884-906 ◽  
Author(s):  
Junichi Omura ◽  
Kimio Satoh ◽  
Nobuhiro Kikuchi ◽  
Taijyu Satoh ◽  
Ryo Kurosawa ◽  
...  

Rationale: Pulmonary arterial hypertension (PAH) is characterized by pulmonary vascular remodeling with aberrant pulmonary artery smooth muscle cells (PASMCs) proliferation, endothelial dysfunction, and extracellular matrix remodeling. Objective: Right ventricular (RV) failure is an important prognostic factor in PAH. Thus, we need to elucidate a novel therapeutic target in both PAH and RV failure. Methods and Results: We performed microarray analysis in PASMCs from patients with PAH (PAH-PASMCs) and controls. We found a ADAMTS8 (disintegrin and metalloproteinase with thrombospondin motifs 8), a secreted protein specifically expressed in the lung and the heart, was upregulated in PAH-PASMCs and the lung in hypoxia-induced pulmonary hypertension (PH) in mice. To elucidate the role of ADAMTS8 in PH, we used vascular smooth muscle cell-specific ADAMTS8-knockout mice (ADAMTS ΔSM22 ). Hypoxia-induced PH was attenuated in ADAMTS ΔSM22 mice compared with controls. ADAMTS8 overexpression increased PASMC proliferation with downregulation of AMPK (AMP-activated protein kinase). In contrast, deletion of ADAMTS8 reduced PASMC proliferation with AMPK upregulation. Moreover, deletion of ADAMTS8 reduced mitochondrial fragmentation under hypoxia in vivo and in vitro. Indeed, PASMCs harvested from ADAMTS ΔSM22 mice demonstrated that phosphorylated DRP-1 (dynamin-related protein 1) at Ser637 was significantly upregulated with higher expression of profusion genes (Mfn1 and Mfn2) and improved mitochondrial function. Moreover, recombinant ADAMTS8 induced endothelial dysfunction and matrix metalloproteinase activation in an autocrine/paracrine manner. Next, to elucidate the role of ADAMTS8 in RV function, we developed a cardiomyocyte-specific ADAMTS8 knockout mice (ADAMTS8 ΔαMHC ). ADAMTS8 ΔαMHC mice showed ameliorated RV failure in response to chronic hypoxia. In addition, ADAMTS8 ΔαMHC mice showed enhanced angiogenesis and reduced RV ischemia and fibrosis. Finally, high-throughput screening revealed that mebendazole, which is used for treatment of parasite infections, reduced ADAMTS8 expression and cell proliferation in PAH-PASMCs and ameliorated PH and RV failure in PH rodent models. Conclusions: These results indicate that ADAMTS8 is a novel therapeutic target in PAH.


2019 ◽  
Vol 40 (Supplement_1) ◽  
Author(s):  
G R T Ryanto ◽  
K Ikeda ◽  
K Miyagawa ◽  
K Yagi ◽  
Y Suzuki ◽  
...  

Abstract Introduction Pulmonary Arterial Hypertension (PAH) is marked by vascular remodeling process that eventually causes pressure increase. Endothelial cells (EC) dysfunction is known to be a major cause for pulmonary vascular remodeling; however, the molecular mechanism remains to be elucidated. Purpose This study aims to identify novel genes and mechanisms involved in PAH development. Methods We performed DNA microarray analysis using RNA samples isolated from human ECs of various vascular beds (including lung microvessels) and organs (including lung). We subsequently searched for genes highly and specifically expressed in lung microvessels since these genes are likely involved in pulmonary circulation homeostasis maintenance. Once found, we confirmed its expressional changes during hypoxia in ECs and lung tissues. We next analyzed its role in EC functions using human pulmonary artery ECs (hPAECs) by in vitro angiogenesis assay, using both candidate gene overexpression via retrovirus transfection and treatment with its active form using appropriate recombinant protein. To explore the role of candidate gene in PAH development in vivo, we generated EC-specific knockout mice and transgenic mice in which the candidate gene is genetically deleted and activated in ECs, respectively. PAH was induced by chronic hypoxia exposure (10% O2- for 3 weeks). Lastly, to explore the underlying mechanisms, we analyzed expressional alterations in possible signaling pathways in ECs that could relate with the effect of the candidate gene. Results From microarray analysis, we identified inhibin Beta-A (INHBA) as a candidate gene that was highly and specifically expressed in human lung microvascular ECs. INHBA homo-dimerization is known to produce activin A (ActA), a TGF-beta superfamily member. Hypoxia exposure caused significant decrease of INHBA mRNA expression in ECs and mouse lung tissues. Both INHBA overexpression and ActA-treatment in hPAECs caused dramatic reduction of their angiogenic capacities (reduced migration and tube formation capability with increased apoptosis). In vivo, EC-specific INHBA overexpressing mice (VEcad-INHBA-TG) showed exacerbated hypoxia-induced PAH, assessed by higher right ventricular systolic pressure (RVSP) and more severely remodeled pulmonary arteries. By contrast, EC-specific INHBA knockout mice (INHBA-floxed/VEcad-Cre-TG) showed significant amelioration of PAH, shown by reduced RVSP and vascular remodeling. Furthermore, we found that INHBA overexpression and ActA-treatment induced a marked reduction of BMPRII, known to play pivotal roles in PAH, in hPAECs by accelerating its lysosomal degradation. Conclusion We identified a novel gene that is crucially involved in PAH development. INHBA and/or ActA negatively regulates EC functions potentially through its BMPRII-altering capability. Gain- and loss-of-function studies in mice revealed that INHBA pathways are promising therapeutic targets for the treatment of PAH.


Sign in / Sign up

Export Citation Format

Share Document