1
|
Chau MHK, Anderson SA, Song R, Cooper L, Ward PA, Yuan B, Shaw C, Stankiewicz P, Cheung SW, Vossaert L, Wang Y, Owen NM, Smith J, Bacino CA, Schulze KV, Bi W. Detection of Clinically Relevant Monogenic Copy-Number Variants by a Comprehensive Genome-Wide Microarray with Exonic Coverage. Clin Chem 2025; 71:141-154. [PMID: 39749505 DOI: 10.1093/clinchem/hvae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/18/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Disease-causing copy-number variants (CNVs) often encompass contiguous genes and can be detected using chromosomal microarray analysis (CMA). Conversely, CNVs affecting single disease-causing genes have historically been challenging to detect due to their small sizes. METHODS A custom comprehensive CMA (Baylor College of Medicine - BCM v11.2) containing 400k probes and featuring exonic coverage for >4200 known or candidate disease-causing genes was utilized for the detection of CNVs at single-exon resolution. CMA results across a consecutive clinical cohort of more than 13 000 patients referred for genetic investigation at Baylor Genetics were examined. The genomic characteristics of CNVs impacting single protein-coding genes were investigated. RESULTS Pathogenic or likely pathogenic (P/LP) CNVs (n = 190) affecting single protein-coding genes were detected in 188 patients, accounting for 9.9% (188/1894) of patients with P/LP CMA findings. The P/LP monogenic CNVs accounted for 9.2% (190/2058) of all P/LP nuclear CNVs detected by CMA. A total of 57.9% (110/190) of P/LP monogenic CNVs were smaller than 50 kb in size. Single exons were affected by 26.3% (50/190) of P/LP monogenic CNVs while 13.2% (25/190) affected 2 exons. CNVs were detected across 107 unique genes associated with predominantly autosomal dominant (AD) and X-linked (XL) conditions but also contributed to autosomal recessive (AR) conditions. CONCLUSIONS CMA with exon-targeted coverage of disease-associated genes facilitated the detection of small CNVs affecting single protein-coding genes, adding substantial clinical sensitivity to comprehensive CNV investigation. This approach resolved monogenic CNVs associated with autosomal and X-linked monogenic etiologies and yielded multiple significant findings. Monogenic CNVs represent an underrecognized subset of disease-causing alleles for Mendelian disorders.
Collapse
Affiliation(s)
- Matthew Hoi Kin Chau
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
- Department of Obstetrics & Gynaecology, The Chinese University of Hong Kong, Hong Kong, China
- The Chinese University of Hong Kong-Baylor College of Medicine Joint Center for Medical Genetics, Hong Kong, China
| | | | - Rodger Song
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Lance Cooper
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Patricia A Ward
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Bo Yuan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Chad Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
| | - Paweł Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Sau Wai Cheung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- The Chinese University of Hong Kong-Baylor College of Medicine Joint Center for Medical Genetics, Hong Kong, China
| | - Liesbeth Vossaert
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Yue Wang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Nichole M Owen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Janice Smith
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Carlos A Bacino
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
- Texas Children's Hospital, Houston, TX, United States
| | - Katharina V Schulze
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| | - Weimin Bi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States
- Baylor Genetics Laboratory, Houston, TX, United States
| |
Collapse
|
2
|
Marinho Y, Villarreal ES, Loya O, Oliveira SD. Mechanisms of lung endothelial cell injury and survival in pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2024; 327:L972-L983. [PMID: 39406383 DOI: 10.1152/ajplung.00208.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 12/06/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, chronic, and incurable inflammatory pulmonary vascular disease characterized by significant sex bias and largely unexplored microbial-associated molecular mechanisms that may influence its development and sex prevalence across various subgroups. PAH can be subclassified as idiopathic, heritable, or associated with conditions such as connective tissue diseases, congenital heart defects, liver disease, infections, and chronic exposure to drugs or toxins. During PAH progression, lung vascular endothelial cells (ECs) undergo dramatic morphofunctional transformations in response to acute and chronic inflammation. These transformations include the appearance and expansion of abnormal vascular cell phenotypes such as those derived from apoptosis-resistant cell growth and endothelial-to-mesenchymal transition (EndoMT). Compelling evidence indicates that these endothelial phenotypes seem to be triggered by chronic lung vascular injury and dysfunction, often characterized by reduced secretion of vasoactive molecules like nitric oxide (NO) and exacerbated response to vasoconstrictors such as Endothelin-1 (ET-1), both long-term known contributors of PAH pathogenesis. This review sheds light on the mechanisms of EC dysfunction, apoptosis, and EndoMT in PAH, aiming to unravel the intricate interactions between ECs, pathogens, and other cell types that drive the onset and progression of this devastating disease. Ultimately, we hope to provide an overview of the complex functions of lung vascular ECs in PAH, inspiring novel therapeutic strategies that target these dysfunctional cells to improve the treatment landscape for PAH, particularly in the face of current and emerging global pathogenic threats.
Collapse
Affiliation(s)
- Ygor Marinho
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
| | - Elizabeth S Villarreal
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
| | - Omar Loya
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
| | - Suellen D Oliveira
- Vascular Immunobiology Lab, Department of Anesthesiology, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, Illinois, United States
| |
Collapse
|
3
|
Wits M, Becher C, de Man F, Sanchez-Duffhues G, Goumans MJ. Sex-biased TGFβ signalling in pulmonary arterial hypertension. Cardiovasc Res 2023; 119:2262-2277. [PMID: 37595264 PMCID: PMC10597641 DOI: 10.1093/cvr/cvad129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare cardiovascular disorder leading to pulmonary hypertension and, often fatal, right heart failure. Sex differences in PAH are evident, which primarily presents with a female predominance and increased male severity. Disturbed signalling of the transforming growth factor-β (TGFβ) family and gene mutations in the bone morphogenetic protein receptor 2 (BMPR2) are risk factors for PAH development, but how sex-specific cues affect the TGFβ family signalling in PAH remains poorly understood. In this review, we aim to explore the sex bias in PAH by examining sex differences in the TGFβ signalling family through mechanistical and translational evidence. Sex hormones including oestrogens, progestogens, and androgens, can determine the expression of receptors (including BMPR2), ligands, and soluble antagonists within the TGFβ family in a tissue-specific manner. Furthermore, sex-related genetic processes, i.e. Y-chromosome expression and X-chromosome inactivation, can influence the TGFβ signalling family at multiple levels. Given the clinical and mechanistical similarities, we expect that the conclusions arising from this review may apply also to hereditary haemorrhagic telangiectasia (HHT), a rare vascular disorder affecting the TGFβ signalling family pathway. In summary, we anticipate that investigating the TGFβ signalling family in a sex-specific manner will contribute to further understand the underlying processes leading to PAH and likely HHT.
Collapse
Affiliation(s)
- Marius Wits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Clarissa Becher
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Frances de Man
- Department of Pulmonary Medicine, Amsterdam University Medical Center (UMC) (Vrije Universiteit), 1081 HV Amsterdam, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
4
|
New Drugs and Therapies in Pulmonary Arterial Hypertension. Int J Mol Sci 2023; 24:ijms24065850. [PMID: 36982922 PMCID: PMC10058689 DOI: 10.3390/ijms24065850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Pulmonary arterial hypertension is a chronic, progressive disorder of the pulmonary vasculature with associated pulmonary and cardiac remodeling. PAH was a uniformly fatal disease until the late 1970s, but with the advent of targeted therapies, the life expectancy of patients with PAH has now considerably improved. Despite these advances, PAH inevitably remains a progressive disease with significant morbidity and mortality. Thus, there is still an unmet need for the development of new drugs and other interventional therapies for the treatment of PAH. One shortcoming of currently approved vasodilator therapies is that they do not target or reverse the underlying pathogenesis of the disease process itself. A large body of evidence has evolved in the past two decades clarifying the role of genetics, dysregulation of growth factors, inflammatory pathways, mitochondrial dysfunction, DNA damage, sex hormones, neurohormonal pathways, and iron deficiency in the pathogenesis of PAH. This review focuses on newer targets and drugs that modify these pathways as well as novel interventional therapies in PAH.
Collapse
|
5
|
Maruyama H, Sakai S, Dewachter L, Dewachter C, Rondelet B, Naeije R, Ieda M. Prostacyclin receptor agonists induce DUSP1 to inhibit pulmonary artery smooth muscle cell proliferation. Life Sci 2023; 315:121372. [PMID: 36608870 DOI: 10.1016/j.lfs.2023.121372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/08/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023]
Abstract
AIMS Upregulated p38MAPK signaling is implicated in the accelerated proliferation of pulmonary artery smooth muscle cells (PA-SMCs) and the pathogenesis of pulmonary artery remodeling observed in pulmonary arterial hypertension (PAH). Previously, we reported that after endothelin-1 (ET-1) pretreatment, bone morphogenetic protein 2 (BMP2) activates p38MAPK signaling and accelerates PA-SMC proliferation. The activity of p38MAPK signaling is tightly regulated by the inactivation of dual-specificity phosphatase 1 (DUSP1). Activated p38MAPK induces DUSP1 expression, forming a negative feedback loop. Prostacyclin IP receptor agonists (prostacyclin and selexipag) are used to treat PAH. In this study, we aimed to verify whether IP receptor agonists affect DUSP1 expression and accelerate the proliferation of PA-SMCs. MAIN METHODS PA-SMCs were treated with BMP2, ET-1, prostacyclin, and MRE-269, an active metabolite of selexipag, either alone or in combination. We quantified mRNA expressions using real-time quantitative polymerase chain reaction. Pulmonary artery specimens and PA-SMCs were obtained during lung transplantation in patients with PAH. KEY FINDINGS Both prostacyclin and MRE-269 increased DUSP1 expression. Combined treatment with BMP2 and ET-1 induced cyclin D1 and DUSP1 expression and increased PA-SMC proliferation. MRE-269 attenuated BMP2/ET-1-induced cell proliferation. ET-1 increased DUSP1 expression in PA-SMCs from control patients but not in PA-SMCs from patients with PAH. SIGNIFICANCE This study showed that the p38MAPK/DUSP1 negative feedback loop is impaired in PAH, contributing to unregulated p38MAPK activation and PA-SMC hyperplasia. IP receptor agonist MRE-269 increases DUSP1 expression and inhibit p38MAPK-mediated PA-SMC proliferation. Future elucidation of the detailed mechanism underlying reduced DUSP1 expression would be informative for PAH treatment.
Collapse
Affiliation(s)
- Hidekazu Maruyama
- Department of Cardiology, National Hospital Organization Kasumigaura Medical Center, 300-8585 Tsuchiura, Japan; Division of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, 305-8577 Tsukuba, Japan; Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium.
| | - Satoshi Sakai
- Faculty of Health Science, Tsukuba University of Technology, 305-8520 Tsukuba, Japan
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium; Department of Cardiology, Erasme Academic Hospital, 1070 Brussels, Belgium
| | - Benoit Rondelet
- Department of Cardiac, Vascular and Thoracic Surgery, CHU UCL Namur, 5530 Yvoir, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, 1070 Brussels, Belgium
| | - Masaki Ieda
- Division of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, 305-8577 Tsukuba, Japan
| |
Collapse
|
6
|
Maruyama H, Sakai S, Ieda M. Endothelin-1 Alters BMP Signaling to Promote Proliferation of Pulmonary Artery Smooth Muscle Cells. Can J Physiol Pharmacol 2022; 100:1018-1027. [PMID: 36037530 DOI: 10.1139/cjpp-2022-0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is characterized by abnormal outgrowth of pulmonary artery smooth muscle cells (PASMCs) of the media. Abundant expression of endothelin-1 (ET-1) and activated p38 mitogen-activated protein kinase (p38MAPK) has been observed in PAH patients. p38MAPK has been implicated in cell proliferation. An unspecified disturbance in bone morphogenetic protein (BMP) signaling may be involved in the development of PAH. Type I receptors (BMPR1A and BMPR1B) and type II receptors (BMPR2) transduce signals via two distinct pathways, i.e., canonical and non-canonical pathways, activating Smad1/5/8 and p38MAPK, respectively. BMPR1B expression was previously reported to be enhanced in the PASMCs of patients with idiopathic PAH. BMP15 binds specifically to BMPR1B. We assessed the effects of ET-1 on BMP receptor expression and cell proliferation. BMP2 increased BMPR1B expression in human PASMCs after pretreatment with ET-1 in vitro. Although BMP2 alone did not affect PASMC proliferation, BMP2 treatment after ET-1 pretreatment significantly accelerated PASMC proliferation. PH-797804, a selective p38MAPK inhibitor, abrogated this proliferation. Similarly, after ET-1 pretreatment, BMP15 significantly accelerated the proliferation of PASMCs, whereas stimulation with BMP15 alone did not. In conclusion, in PASMCs, ET-1 exposure under pathological conditions alters BMP signaling to activate p38MAPK, resulting in cell proliferation.
Collapse
Affiliation(s)
- Hidekazu Maruyama
- National Hospital Organisation Kasumigaura Medical Center Internal Medicine, Cardiology, Tsuchiura, Japan;
| | - Satoshi Sakai
- University of Tsukuba Faculty of Medicine, Tsukuba, Ibaraki, Japan;
| | - Masaki Ieda
- University of Tsukuba Faculty of Medicine, Tsukuba, Ibaraki, Japan;
| |
Collapse
|
7
|
King NE, Brittain E. Emerging therapies: The potential roles SGLT2 inhibitors, GLP1 agonists, and ARNI therapy for ARNI pulmonary hypertension. Pulm Circ 2022; 12:e12028. [PMID: 35506082 PMCID: PMC9052991 DOI: 10.1002/pul2.12028] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary hypertension (PH) is a highly morbid condition. PH due to left heart disease (PH-LHD) has no specific therapies and pulmonary arterial hypertension (PAH) has substantial residual risk despite several approved therapies. Multiple lines of experimental evidence link metabolic dysfunction to the pathogenesis and outcomes in PH-LHD and PAH, and novel metabolic agents hold promise to improve outcomes in these populations. The antidiabetic sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide-1 (GLP1) agonists targeting metabolic dysfunction and improve outcomes in patients with LHD but have not been tested specifically in patients with PH. The angiotensin receptor/neprilysin inhibitors (ARNIs) produce significant improvements in cardiac hemodynamics and may improve metabolic dysfunction that could benefit the pulmonary circulation and right ventricle function. On the basis of promising preclinical work with these medications and clinical rationale, we explore the potential of SGLT2 inhibitors, GLP1 agonists, and ARNIs as therapies for both PH-LHD and PAH.
Collapse
Affiliation(s)
| | - Evan Brittain
- Department of Medicine, Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
8
|
Xin Z, Wang J, Li S, Sun C, Jiang W, Xin Q, Wang J, Qi T, Li K, Zhang Z, Luan Y. A review of BMP and Wnt signaling pathway in the pathogenesis of pulmonary arterial hypertension. Clin Exp Hypertens 2021; 44:175-180. [PMID: 34821188 DOI: 10.1080/10641963.2021.1996590] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a chronic disease characterized by a progressive elevation in mean pulmonary arterial pressure. This occurs due to abnormal remodeling of small peripheral lung vasculature resulting in progressive occlusion of the artery lumen that eventually causes right heart failure and death. Current therapeutic options for PAH are limited and focused mainly on reversal of pulmonary vasoconstriction and proliferation of vascular cells. Although these treatments can relieve disease symptoms, PAH remains a progressive lethal disease.Bone morphogenetic proteins (BMPs) and their receptors were required for PAH-induced right ventricular hypertrophy. Emerging data suggest that restoration of BMP type II receptor (BMPR2) signaling in PAH is a promising alternative that could prevent and reverse pulmonary vascular remodeling. BMPR2 mutations have been identified in >70% of familial and roughly 15% of sporadic PAH cases. Wingless (Wnt) are a family of secreted glycoproteins with varying expression patterns and a range of functions, Wnt signaling pathway is divided into canonical signaling pathway and non-canonical signaling pathway. A recent study reports that interaction between BMP and Wnt closely associated with lung development, those cascade coordination regulation stem cell fate which determine lung branching morphogenes. The promoting effect of BMPR2 on proliferation, survival, and motility of endothelial cells was through recruiting Wnts signaling pathway, the interaction between BMP and Wnt closely associated with lung development.Therefore, in this review, we outline the latest advances of BMP and Wnt signaling pathway in the pathogenesis of PAH and disease progression.
Collapse
Affiliation(s)
- Zhihong Xin
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University
| | - Junfu Wang
- Clinical laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University
| | - Susu Li
- College of pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, 271000, China
| | - Chao Sun
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Wan Jiang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Qian Xin
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Jue Wang
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Tonggnag Qi
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Kailin Li
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| | - Zhaohua Zhang
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University
| | - Yun Luan
- Institute of Medical Science, The Second Hospital, Cheeloo College of Medicine, Shandong University, No. 247, Beiyuan Dajie, Jinan, 250033, P.R. China
| |
Collapse
|
9
|
Ion channels as convergence points in the pathology of pulmonary arterial hypertension. Biochem Soc Trans 2021; 49:1855-1865. [PMID: 34346486 PMCID: PMC8421048 DOI: 10.1042/bst20210538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease of the cardiopulmonary system that lacks curative treatments. The main pathological event in PAH is elevated vascular resistance in the pulmonary circulation, caused by abnormal vasoconstriction and vascular remodelling. Ion channels are key determinants of vascular smooth muscle tone and homeostasis, and four PAH channelopathies (KCNK3, ABCC8, KCNA5, TRPC6) have been identified so far. However, the contribution of ion channels in other forms of PAH, which account for the majority of PAH patients, has been less well characterised. Here we reason that a variety of triggers of PAH (e.g. BMPR2 mutations, hypoxia, anorectic drugs) that impact channel function may contribute to the onset of the disease. We review the molecular mechanisms by which these ‘extrinsic’ factors converge on ion channels and provoke their dysregulation to promote the development of PAH. Ion channels of the pulmonary vasculature are therefore promising therapeutic targets because of the modulation they provide to both vasomotor tone and proliferation of arterial smooth muscle cells.
Collapse
|
10
|
Novel Genetic and Molecular Pathways in Pulmonary Arterial Hypertension Associated with Connective Tissue Disease. Cells 2021; 10:cells10061488. [PMID: 34199176 PMCID: PMC8231632 DOI: 10.3390/cells10061488] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022] Open
Abstract
Pulmonary Arterial Hypertension (PAH) is a severe complication of Connective Tissue Disease (CTD), with remarkable morbidity and mortality. However, the molecular and genetic basis of CTD-PAH remains incompletely understood. This study aimed to screen for genetic defects in a cohort of patients with CTD-PAH, using a PAH-specific panel of 35 genes. During recruitment, 79 patients were studied, including 59 Systemic Sclerosis patients (SSc) and 69 females. Disease-associated variants were observed in nine patients: 4 pathogenic/likely pathogenic variants in 4 different genes (TBX4, ABCC8, KCNA5 and GDF2/BMP9) and 5 Variants of Unknown Significance (VUS) in 4 genes (ABCC8, NOTCH3, TOPBP1 and CTCFL). One patient with mixed CTD had a frameshift pathogenic variant in TBX4. Two patients with SSc-PAH carried variants in ABCC8. A patient diagnosed with Systemic Lupus Erythematous (SLE) presented a pathogenic nonsense variant in GDF2/BMP9. Another patient with SSc-PAH presented a pathogenic variant in KCNA5. Four patients with SSc-PAH carried a VUS in NOTCH1, CTCFL, CTCFL and TOPBP1, respectively. These findings suggest that genetic factors may contribute to Pulmonary Vascular Disease (PVD) in CTD patients.
Collapse
|
11
|
Gorelova A, Berman M, Al Ghouleh I. Endothelial-to-Mesenchymal Transition in Pulmonary Arterial Hypertension. Antioxid Redox Signal 2021; 34:891-914. [PMID: 32746619 PMCID: PMC8035923 DOI: 10.1089/ars.2020.8169] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/14/2022]
Abstract
Endothelial-to-mesenchymal transition (EndMT) is a process that encompasses extensive transcriptional reprogramming of activated endothelial cells leading to a shift toward mesenchymal cellular phenotypes and functional responses. Initially observed in the context of embryonic development, in the last few decades EndMT is increasingly recognized as a process that contributes to a variety of pathologies in the adult organism. Within the settings of cardiovascular biology, EndMT plays a role in various diseases, including atherosclerosis, heart valvular disease, cardiac fibrosis, and myocardial infarction. EndMT is also being progressively implicated in development and progression of pulmonary hypertension (PH) and pulmonary arterial hypertension (PAH). This review covers the current knowledge about EndMT in PH and PAH, and provides comprehensive overview of seminal discoveries. Topics covered include evidence linking EndMT to factors associated with PAH development, including hypoxia responses, inflammation, dysregulation of bone-morphogenetic protein receptor 2 (BMPR2), and redox signaling. This review amalgamates these discoveries into potential insights for the identification of underlying mechanisms driving EndMT in PH and PAH, and discusses future directions for EndMT-based therapeutic strategies in disease management.
Collapse
Affiliation(s)
- Anastasia Gorelova
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mariah Berman
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Imad Al Ghouleh
- Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Puckett DL, Alquraishi M, Chowanadisai W, Bettaieb A. The Role of PKM2 in Metabolic Reprogramming: Insights into the Regulatory Roles of Non-Coding RNAs. Int J Mol Sci 2021; 22:1171. [PMID: 33503959 PMCID: PMC7865720 DOI: 10.3390/ijms22031171] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 01/17/2023] Open
Abstract
Pyruvate kinase is a key regulator in glycolysis through the conversion of phosphoenolpyruvate (PEP) into pyruvate. Pyruvate kinase exists in various isoforms that can exhibit diverse biological functions and outcomes. The pyruvate kinase isoenzyme type M2 (PKM2) controls cell progression and survival through the regulation of key signaling pathways. In cancer cells, the dimer form of PKM2 predominates and plays an integral role in cancer metabolism. This predominance of the inactive dimeric form promotes the accumulation of phosphometabolites, allowing cancer cells to engage in high levels of synthetic processing to enhance their proliferative capacity. PKM2 has been recognized for its role in regulating gene expression and transcription factors critical for health and disease. This role enables PKM2 to exert profound regulatory effects that promote cancer cell metabolism, proliferation, and migration. In addition to its role in cancer, PKM2 regulates aspects essential to cellular homeostasis in non-cancer tissues and, in some cases, promotes tissue-specific pathways in health and diseases. In pursuit of understanding the diverse tissue-specific roles of PKM2, investigations targeting tissues such as the kidney, liver, adipose, and pancreas have been conducted. Findings from these studies enhance our understanding of PKM2 functions in various diseases beyond cancer. Therefore, there is substantial interest in PKM2 modulation as a potential therapeutic target for the treatment of multiple conditions. Indeed, a vast plethora of research has focused on identifying therapeutic strategies for targeting PKM2. Recently, targeting PKM2 through its regulatory microRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) has gathered increasing interest. Thus, the goal of this review is to highlight recent advancements in PKM2 research, with a focus on PKM2 regulatory microRNAs and lncRNAs and their subsequent physiological significance.
Collapse
Affiliation(s)
- Dexter L. Puckett
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Mohammed Alquraishi
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| | - Winyoo Chowanadisai
- Department of Nutrition, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Ahmed Bettaieb
- Department of Nutrition, University of Tennessee Knoxville, Knoxville, TN 37996, USA; (D.L.P.); (M.A.)
| |
Collapse
|
13
|
Maruyama H, Sakai S, Dewachter L, Dewachter C, Rondelet B, Naeije R, Ieda M. Endothelin-1 induces lysyl oxidase expression in pulmonary artery smooth muscle cells. Can J Physiol Pharmacol 2020; 98:629-636. [PMID: 32615041 DOI: 10.1139/cjpp-2019-0658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The increase in thickening of the arterial wall of pulmonary arterial hypertension (PAH) includes cellular proliferation as well as matrix deposition and interrupted internal elastic lamina (IEL) consisting of a thick homogeneous sheet of elastin. Little is, although, known about the detail of IEL formation in PAH. Endothelin-1 is overexpressed in pulmonary arterioles of PAH. We aimed to examine the expression of genes contributing to IEL formation in pulmonary artery smooth muscle cells (PASMCs) especially focused on lysyl oxidase (LOx), an exreacellular matrix enzyme that catalyzes the cross-linking of collagens or elastin. We quantified mRNA expressions of genes contributing to IEL formation including LOx in PASMCs using real-time quantitative polymerase chain reaction. We stimulated human PASMCs with endothelin-1 with prostacyclin or trapidil. Endothelin-1 significantly increased LOx expression. Prostacyclin and trapidil restored endothelin-1-induced LOx expression to the basal level. Endothelin-1 increased LOx expression strongly in PASMCs from PAH patients compared to those from controls. Trapidil reduced LOx expression only in PASMCs from PAH patients. Overexpressed endothelin-1 in PAH patients can increase expression of LOx and agitate cross-linking of elastin and collagen, resulting in ectopic deposition of these in the vascular media.
Collapse
Affiliation(s)
- Hidekazu Maruyama
- Department of Cardiology, National Hospital Organization Kasumigaura Medical Center, Tsuchiura, Japan.,Faculty of Health Science, Tsukuba University of Technology, Tsukuba, Japan.,Division of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Satoshi Sakai
- Faculty of Health Science, Tsukuba University of Technology, Tsukuba, Japan.,Division of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium.,Department of Cardiology, Erasme Academic Hospital, Brussels, Belgium
| | - Benoit Rondelet
- Department of Cardiac, Vascular and Thoracic Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Masaki Ieda
- Division of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
14
|
The BMP Receptor 2 in Pulmonary Arterial Hypertension: When and Where the Animal Model Matches the Patient. Cells 2020; 9:cells9061422. [PMID: 32521690 PMCID: PMC7348993 DOI: 10.3390/cells9061422] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/30/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022] Open
Abstract
Background: Mutations in bone morphogenetic protein receptor type II (BMPR2) are leading to the development of hereditary pulmonary arterial hypertension (PAH). In non-hereditary forms of PAH, perturbations in the transforming growth factor-β (TGF-β)/BMP-axis are believed to cause deficient BMPR2 signaling by changes in receptor expression, the activity of the receptor and/or downstream signaling. To date, BMPR2 expression and its activity in the lungs of patients with non-hereditary PAH is poorly characterized. In recent decades, different animal models have been used to understand the role of BMPR2 signaling in PAH pathophysiology. Specifically, the monocrotaline (MCT) and Sugen-Hypoxia (SuHx) models are extensively used in interventional studies to examine if restoring BMPR2 signaling results in PAH disease reversal. While PAH is assumed to develop in patients over months or years, pulmonary hypertension in experimental animal models develops in days or weeks. It is therefore likely that modifications in BMP and TGF-β signaling in these models do not fully recapitulate those in patients. In order to determine the translational potential of the MCT and SuHx models, we analyzed the BMPR2 expression and activity in the lungs of rats with experimentally induced PAH and compared this to the BMPR2 expression and activity in the lungs of PAH patients. Methods: the BMPR2 expression was analyzed by Western blot analysis and immunofluorescence (IF) microscopy to determine the quantity and localization of the receptor in the lung tissue from normal control subjects and patients with hereditary or idiopathic PAH, as well as in the lungs of control rats and rats with MCT or SuHx-induced PAH. The activation of the BMP pathway was analyzed by determining the level and localization of phosphorylated Smad1/5/8 (pSmad 1/5/8), a downstream mediator of canonical BMPR2 signaling. Results: While BMPR2 and pSmad 1/5/8 expression levels were unaltered in whole lung lysates/homogenates from patients with hereditary and idiopathic PAH, IF analysis showed that BMPR2 and pSmad 1/5/8 levels were markedly decreased in the pulmonary vessels of both PAH patient groups. Whole lung BMPR2 expression was variable in the two PAH rat models, while in both experimental models the expression of BMPR2 in the lung vasculature was increased. However, in the human PAH lungs, the expression of pSmad 1/5/8 was downregulated in the lung vasculature of both experimental models. Conclusion: BMPR2 receptor expression and downstream signaling is reduced in the lung vasculature of patients with idiopathic and hereditary PAH, which cannot be appreciated when using human whole lung lysates. Despite increased BMPR2 expression in the lung vasculature, the MCT and SuHx rat models did develop PAH and impaired downstream BMPR2-Smad signaling similar to our findings in the human lung.
Collapse
|
15
|
Hemnes AR, Fessel JP, Chen X, Zhu S, Fortune NL, Jetter C, Freeman M, Newman JH, West JD, Talati MH. BMPR2 dysfunction impairs insulin signaling and glucose homeostasis in cardiomyocytes. Am J Physiol Lung Cell Mol Physiol 2020; 318:L429-L441. [PMID: 31850803 PMCID: PMC7052666 DOI: 10.1152/ajplung.00555.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 11/04/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022] Open
Abstract
Insulin resistance and right ventricular (RV) dysfunction are associated with lipotoxicity in heritable forms of pulmonary arterial hypertension (PAH), commonly due to mutations in bone morphogenetic protein receptor type 2 (BMPR2). How BMPR2 dysfunction in cardiomyocytes alters glucose metabolism and the response of these cells to insulin are unknown. We hypothesized that BMPR2 mutation in cardiomyocytes alters glucose-supported mitochondrial respiration and impairs cellular responses to insulin, including glucose and lipid uptake. We performed metabolic assays, immunofluorescence and Western analysis, RNA profiling, and radioactive isotope uptake studies in H9c2 cardiomyocyte cell lines with and without patient-derived BMPR2 mutations (mutant cells), with and without insulin. Unlike control cells, BMPR2 mutant cardiomyocytes have reduced metabolic plasticity as indicated by reduced mitochondrial respiration with increased mitochondrial superoxide production. These mutant cells show enhanced baseline phosphorylation of insulin-signaling protein as indicated by increased Akt, AMPK, and acetyl-CoA carboxylase phosphorylation that may negatively influence fatty acid oxidation and enhance lipid uptake, and are insulin insensitive. Furthermore, mutant cells demonstrate an increase in milk fat globule-EGF factor-8 protein (MFGE8), which influences the insulin-signaling pathway by phosphorylating AktSer473 via phosphatidylinositol 3-kinase and mammalian target of rapamycin. In conclusion, BMPR2 mutant cardiomyocytes have reduced metabolic plasticity and fail to respond to glucose. These cells have enhanced baseline insulin-signaling pattern favoring insulin resistance with failure to augment this pattern in response to insulin. BMPR2 mutation possibly blunts glucose uptake and enhances lipid uptake in these cardiomyocytes. The MFGE8-driven signaling pathway may suggest a new mechanism underlying RV lipotoxicity in PAH.
Collapse
Affiliation(s)
- Anna R Hemnes
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Joshua P Fessel
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Xinping Chen
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Shijun Zhu
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Niki L Fortune
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Christopher Jetter
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Michael Freeman
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - John H Newman
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James D West
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Megha H Talati
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
16
|
Comeglio P, Filippi S, Sarchielli E, Morelli A, Cellai I, Corno C, Adorini L, Vannelli GB, Maggi M, Vignozzi L. Therapeutic effects of the selective farnesoid X receptor agonist obeticholic acid in a monocrotaline-induced pulmonary hypertension rat model. J Endocrinol Invest 2019; 42:951-965. [PMID: 30674010 DOI: 10.1007/s40618-019-1009-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/11/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Activation of the farnesoid X receptor (FXR), a member of the nuclear receptor steroid superfamily, leads to anti-inflammatory and anti-fibrotic effects in several tissues, including the lung. We have recently demonstrated a protective effect of the farnesoid X receptor (FXR) agonist obeticholic acid (OCA) in rat models of monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) and bleomycin-induced pulmonary fibrosis. The aim of the present study was to investigate whether the positive effects of OCA treatment could be exerted also in established MCT-induced PAH, i.e., starting treatment 2 weeks after MCT administration. METHODS Rats with MCT-induced PAH were treated, 2 weeks after MCT administration, with OCA or tadalafil for two additional weeks. Pulmonary functional tests were performed at week 2 (before treatment) and four (end of treatment). At the same time points, lung morphological features and expression profile of genes related to smooth muscle relaxation/contraction and tissue remodeling were also assessed. RESULTS 2 weeks after MCT-induced injury, the treadmill resistance (a functional parameter related to pulmonary hypertension) was significantly decreased. At the same time point, we observed right ventricular hypertrophy and vascular remodeling, with upregulation of genes related to inflammation. At week 4, we observed a further worsening of the functional and morphological parameters, accompanied by dysregulation of inflammatory and extracellular matrix markers mRNA expression. Administration of OCA (3 or 10 mg/kg/day), starting 2 weeks after MCT-induced injury, significantly improved pulmonary function, effectively normalizing the exercise capacity. OCA also reverted most of the lung alterations, with a significant reduction of lung vascular wall thickness, right ventricular hypertrophy, and restoration of the local balance between relaxant and contractile pathways. Markers of remodeling pathways were also normalized by OCA treatment. Notably, results with OCA treatment were similar, or even superior, to those obtained with tadalafil, a recently approved treatment for pulmonary hypertension. CONCLUSIONS The results of this study demonstrate a significant therapeutic effect of OCA in established MCT-induced PAH, improving exercise capacity associated with reduction of right ventricular hypertrophy and lung vascular remodeling. Thus, OCA dosing in a therapeutic protocol restores the balance between relaxant and contractile pathways in the lung, promoting cardiopulmonary protective actions in MCT-induced PAH.
Collapse
Affiliation(s)
- P Comeglio
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - S Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of Reproduction, Department of NEUROFARBA, University of Florence, Florence, Italy
| | - E Sarchielli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - A Morelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - I Cellai
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - C Corno
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - L Adorini
- Intercept Pharmaceuticals, New York, NY, USA
| | - G B Vannelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - M Maggi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
- I.N.B.B. (Istituto Nazionale Biostrutture E Biosistemi), Rome, Italy
| | - L Vignozzi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy.
- I.N.B.B. (Istituto Nazionale Biostrutture E Biosistemi), Rome, Italy.
| |
Collapse
|
17
|
Abbasi Y, Jabbari J, Jabbari R, Glinge C, Izadyar S, Spiekerkoetter E, Zamanian RT, Carlsen J, Tfelt‐Hansen J. Exome data clouds the pathogenicity of genetic variants in Pulmonary Arterial Hypertension. Mol Genet Genomic Med 2018; 6:835-844. [PMID: 30084161 PMCID: PMC6160702 DOI: 10.1002/mgg3.452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 04/25/2018] [Accepted: 06/03/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We aimed to provide a set of previously reported PAH-associated missense and nonsense variants, and evaluate the pathogenicity of those variants. METHODS The Human Gene Mutation Database, PubMed, and Google Scholar were searched for previously reported PAH-associated genes and variants. Thereafter, both exome sequencing project and exome aggregation consortium as background population searched for previously reported PAH-associated missense and nonsense variants. The pathogenicity of previously reported PAH-associated missense variants evaluated by using four in silico prediction tools. RESULTS In total, 14 PAH-associated genes and 180 missense and nonsense variants were gathered. The BMPR2, the most frequent reported gene, encompasses 135 of 180 missense and nonsense variants. The exome sequencing project comprised 9, and the exome aggregation consortium counted 25 of 180 PAH-associated missense and nonsense variants. The TOPBP1 and ENG genes are unlikely to be the monogenic cause of PAH pathogenesis based on allele frequency in background population and prediction analysis. CONCLUSION This is the first evaluation of previously reported PAH-associated missense and nonsense variants. The BMPR2 identified as the major gene out of 14 PAH-associated genes. Based on findings, the ENG and TOPBP1 gene are not likely to be the monogenic cause of PAH.
Collapse
Affiliation(s)
- Yeganeh Abbasi
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | | | - Reza Jabbari
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Charlotte Glinge
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Seyed Bahador Izadyar
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical CareStanford University School of MedicineCalifornia
| | - Roham T. Zamanian
- Division of Pulmonary and Critical CareStanford University School of MedicineCalifornia
| | - Jørn Carlsen
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
| | - Jacob Tfelt‐Hansen
- Heart CentreDepartment of CardiologyCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of CardiologySection for Pulmonary Hypertension and Right Heart FailureCopenhagen University Hospital, RigshospitaletCopenhagenDenmark
- Department of Forensic MedicineFaculty of Medical SciencesUniversity of CopenhagenDenmark
| |
Collapse
|
18
|
Caruso P, Dunmore BJ, Schlosser K, Schoors S, Dos Santos C, Perez-Iratxeta C, Lavoie JR, Zhang H, Long L, Flockton AR, Frid MG, Upton PD, D'Alessandro A, Hadinnapola C, Kiskin FN, Taha M, Hurst LA, Ormiston ML, Hata A, Stenmark KR, Carmeliet P, Stewart DJ, Morrell NW. Identification of MicroRNA-124 as a Major Regulator of Enhanced Endothelial Cell Glycolysis in Pulmonary Arterial Hypertension via PTBP1 (Polypyrimidine Tract Binding Protein) and Pyruvate Kinase M2. Circulation 2017; 136:2451-2467. [PMID: 28971999 DOI: 10.1161/circulationaha.117.028034] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 09/08/2017] [Indexed: 01/24/2023]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized by abnormal growth and enhanced glycolysis of pulmonary artery endothelial cells. However, the mechanisms underlying alterations in energy production have not been identified. METHODS Here, we examined the miRNA and proteomic profiles of blood outgrowth endothelial cells (BOECs) from patients with heritable PAH caused by mutations in the bone morphogenetic protein receptor type 2 (BMPR2) gene and patients with idiopathic PAH to determine mechanisms underlying abnormal endothelial glycolysis. We hypothesized that in BOECs from patients with PAH, the downregulation of microRNA-124 (miR-124), determined with a tiered systems biology approach, is responsible for increased expression of the splicing factor PTBP1 (polypyrimidine tract binding protein), resulting in alternative splicing of pyruvate kinase muscle isoforms 1 and 2 (PKM1 and 2) and consequently increased PKM2 expression. We questioned whether this alternative regulation plays a critical role in the hyperglycolytic phenotype of PAH endothelial cells. RESULTS Heritable PAH and idiopathic PAH BOECs recapitulated the metabolic abnormalities observed in pulmonary artery endothelial cells from patients with idiopathic PAH, confirming a switch from oxidative phosphorylation to aerobic glycolysis. Overexpression of miR-124 or siRNA silencing of PTPB1 restored normal proliferation and glycolysis in heritable PAH BOECs, corrected the dysregulation of glycolytic genes and lactate production, and partially restored mitochondrial respiration. BMPR2 knockdown in control BOECs reduced the expression of miR-124, increased PTPB1, and enhanced glycolysis. Moreover, we observed reduced miR-124, increased PTPB1 and PKM2 expression, and significant dysregulation of glycolytic genes in the rat SUGEN-hypoxia model of severe PAH, characterized by reduced BMPR2 expression and endothelial hyperproliferation, supporting the relevance of this mechanism in vivo. CONCLUSIONS Pulmonary vascular and circulating progenitor endothelial cells isolated from patients with PAH demonstrate downregulation of miR-124, leading to the metabolic and proliferative abnormalities in PAH ECs via PTPB1 and PKM1/PKM2. Therefore, the manipulation of this miRNA or its targets could represent a novel therapeutic approach for the treatment of PAH.
Collapse
Affiliation(s)
- Paola Caruso
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Benjamin J Dunmore
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Kenny Schlosser
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Sandra Schoors
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (S.S., P.C.).,Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Belgium (S.S., P.C.)
| | - Claudia Dos Santos
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.S., M.L.O.)
| | - Carol Perez-Iratxeta
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Jessie R Lavoie
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Hui Zhang
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Lu Long
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Amanda R Flockton
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Maria G Frid
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Paul D Upton
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | | | - Charaka Hadinnapola
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Fedir N Kiskin
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Mohamad Taha
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Liam A Hurst
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| | - Mark L Ormiston
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada (C.D.S., M.L.O.)
| | - Akiko Hata
- University of Colorado, Anschutz Medical Campus, Aurora. Cardiovascular Research Institute, University of California, San Francisco (A.H.)
| | - Kurt R Stenmark
- Cardiovascular Pulmonary Research Laboratories, Departments of Pediatrics and Medicine (H.Z., A.R.F., M.G.F., K.R.S.)
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, VIB, Leuven, Belgium (S.S., P.C.).,Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Belgium (S.S., P.C.)
| | - Duncan J Stewart
- Ottawa Hospital Research Institute and University of Ottawa, Ontario, Canada (K.S., C.P.-I., J.R.L., M.T., D.J.S.)
| | - Nicholas W Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's Hospital, United Kingdom (P.C., B.J.D., L.L., P.D.U., C.H., F.N.K., L.A.H.., N.W.M.)
| |
Collapse
|
19
|
|
20
|
Zhang Z, Zhang L, Sun C, Kong F, Wang J, Xin Q, Jiang W, Li K, Chen O, Luan Y. Baicalin attenuates monocrotaline-induced pulmonary hypertension through bone morphogenetic protein signaling pathway. Oncotarget 2017; 8:63430-63441. [PMID: 28969002 PMCID: PMC5609934 DOI: 10.18632/oncotarget.18825] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/01/2017] [Indexed: 11/25/2022] Open
Abstract
Baicalin, a flavonoid compound extracted from roots of Scutellaria baicalensis Georgi (huang qin), it has been shown to effectively attenuates pulmonary hypertension (PH), however, the potential mechanism remains unexplored. In this study, we investigated the potential mechanism of baicalin on monocrotaline (MCT)-induced PH in rats. The results showed that baicalin attenuated lung damage in PH rat model through inhibiting the pulmonary arterial smooth muscle cell proliferation and induction of cells apoptosis. Furthermore, we demonstrated that baicalin inhibition the expression of nuclear factor-κB (NF-κB) p65 and bone morphogenetic protein (BMP) antagonists gremlin-1, but increased the expression of inhibitor of NF-κB (I-κBα), BMPR2, BMP-4, BMP-9 and Smad1/5/8. Additionally, baicalin suppression endothelial-to-mesenchymal transition in PH lung tissue. Collectively, we confirmed that baicalin via inhibition of NF-κB signaling to further activation of BMP signaling to have a therapeutic effect on PH and providing a promising therapeutic strategy for PH.
Collapse
Affiliation(s)
- Zhaohua Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Luan Zhang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, China
| | - Chao Sun
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Feng Kong
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Jue Wang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Qian Xin
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Wen Jiang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Kaili Li
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Ou Chen
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Yun Luan
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
21
|
Sankhe S, Manousakidi S, Antigny F, Arthur Ataam J, Bentebbal S, Ruchon Y, Lecerf F, Sabourin J, Price L, Fadel E, Dorfmüller P, Eddahibi S, Humbert M, Perros F, Capuano V. T-type Ca 2+ channels elicit pro-proliferative and anti-apoptotic responses through impaired PP2A/Akt1 signaling in PASMCs from patients with pulmonary arterial hypertension. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1631-1641. [PMID: 28655554 DOI: 10.1016/j.bbamcr.2017.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/13/2017] [Accepted: 06/21/2017] [Indexed: 10/19/2022]
Abstract
Idiopathic pulmonary arterial hypertension (iPAH) is characterized by obstructive hyperproliferation and apoptosis resistance of distal pulmonary artery smooth muscle cells (PASMCs). T-type Ca2+ channel blockers have been shown to reduce experimental pulmonary hypertension, although the impact of T-type channel inhibition remains unexplored in PASMCs from iPAH patients. Here we show that T-type channels Cav3.1 and Cav3.2 are present in the lung and PASMCs from iPAH patients and control subjects. The blockade of T-type channels by the specific blocker, TTA-A2, prevents cell cycle progression and PASMCs growth. In iPAH cells, T-type channel signaling fails to activate phosphatase PP2A, leading to an increase in ERK1/2, P38 activation. Moreover, T-type channel signaling is redirected towards the activation of the kinase Akt1, leading to increased expression of the anti-apoptotic protein survivin, and a decrease in the pro-apoptotic mediator FoxO3A. Finally, in iPAH cells, Akt1 is no longer able to regulate caspase 9 activation, whereas T-type channel overexpression reverses PP2A defect in iPAH cells but reinforces the deleterious effects of Akt1 activation. Altogether, these data highlight T-type channel signaling as a strong trigger of the pathological phenotype of PASMCs from iPAH patients (hyper-proliferation/cells survival and apoptosis resistance), suggesting that both T-type channels and PP2A may be promising therapeutic targets for pulmonary hypertension.
Collapse
Affiliation(s)
- Safietou Sankhe
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Sevasti Manousakidi
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Fabrice Antigny
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jennifer Arthur Ataam
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Sana Bentebbal
- PhyMedExp, Univ. Montpellier, Inserm U1046, cNRS UMR9214.34295 MINSERM U1046, Montpellier, France
| | - Yann Ruchon
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Florence Lecerf
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jessica Sabourin
- INSERM UMR-S1180, Univ. Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Laura Price
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK
| | - Elie Fadel
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Peter Dorfmüller
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Saadia Eddahibi
- PhyMedExp, Univ. Montpellier, Inserm U1046, cNRS UMR9214.34295 MINSERM U1046, Montpellier, France
| | - Marc Humbert
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France; AP-HP, Service de pneumologie, Hôpital Bicêtre, Le Kremlin Bicêtre, France
| | - Frédéric Perros
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France
| | - Véronique Capuano
- INSERM U999, Hôpital Marie Lannelongue, Le Plessis Robinson, France; Univ. Paris-Sud, Faculté de Médecine, Univ. Paris-Saclay, Le Kremlin Bicêtre, France.
| |
Collapse
|
22
|
Quarck R, Perros F. Rescuing BMPR2-driven endothelial dysfunction in PAH: a novel treatment strategy for the future? Stem Cell Investig 2017; 4:56. [PMID: 28725652 DOI: 10.21037/sci.2017.05.11] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 05/15/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Rozenn Quarck
- Respiratory Division, University Hospitals and Department of Clinical and Experimental Medicine, University of Leuven, Leuven, Belgium
| | - Fréderic Perros
- INSERM UMR_S 999, Centre Chirurgical Marie Lannelongue, Université Paris-Sud, Le Plessis-Robinson, France
| |
Collapse
|
23
|
Cai P, Kovacs L, Dong S, Wu G, Su Y. BMP4 inhibits PDGF-induced proliferation and collagen synthesis via PKA-mediated inhibition of calpain-2 in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L638-L648. [PMID: 28235949 PMCID: PMC5451598 DOI: 10.1152/ajplung.00260.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 02/09/2017] [Accepted: 02/23/2017] [Indexed: 11/22/2022] Open
Abstract
In the present study, we investigated the effect of bone morphogenetic protein 4 (BMP4) on PDGF-induced cell proliferation and collagen synthesis in pulmonary artery smooth muscle cells (PASMCs). Normal human PASMCs were incubated with and without PDGF-BB in the absence and presence of BMP4 for 0.5 to 24 h. The protein levels of collagen-I, p-Smad2/3, p-Smad1/5, and intracellular active TGF-β1, calpain activity, and cell proliferation were then measured. The results showed that BMP4 induced an increase in p-Smad1/5 but had no effect on the protein levels of collagen-I, p-Smad2/3, and intracellular active TGF-β1 and calpain activity in control PASMCs. Nevertheless, BMP4 attenuated increases in cell proliferation and protein levels of collagen-I, p-Smad2/3, and intracellular active TGF-β1 and calpain activity in PASMCs exposed to PDGF-BB. Moreover, BMP4 increased PKA activity and inhibition of PKA prevented the inhibitory effects of BMP4 on PDGF-BB-induced calpain activation in normal PASMCs. The PKA activator forskolin recapitulated the suppressive effect of BMP4 on PDGF-induced calpain activation. Furthermore, BMP4 prevented a PDGF-induced decrease in calpain-2 phosphorylation at serine-369 in normal PASMCs. Finally, BMP4 did not attenuate PDGF-induced increases in cell proliferation, collagen-I protein levels, and calpain activation and did not induce PKA activation and did not prevent a PDGF-induced decrease in calpain-2 phosphorylation at serine-369 in PASMCs from idiopathic pulmonary arterial hypertension (PAH) patients. These data demonstrate that BMP4 inhibits PDGF-induced cell proliferation and collagen synthesis via PKA-mediated inhibition of calpain-2 in normal PASMCs. The inhibitory effects of BMP4 on PDGF-induced cell proliferation, collagen synthesis, and calpain-2 activation are impaired in PASMCs from PAH patients, which may contribute to pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Pengcheng Cai
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Laszlo Kovacs
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Sam Dong
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia; .,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia.,Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Research Service, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia; and
| |
Collapse
|
24
|
TNFα drives pulmonary arterial hypertension by suppressing the BMP type-II receptor and altering NOTCH signalling. Nat Commun 2017; 8:14079. [PMID: 28084316 PMCID: PMC5241886 DOI: 10.1038/ncomms14079] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 11/28/2016] [Indexed: 02/08/2023] Open
Abstract
Heterozygous germ-line mutations in the bone morphogenetic protein type-II receptor (BMPR-II) gene underlie heritable pulmonary arterial hypertension (HPAH). Although inflammation promotes PAH, the mechanisms by which inflammation and BMPR-II dysfunction conspire to cause disease remain unknown. Here we identify that tumour necrosis factor-α (TNFα) selectively reduces BMPR-II transcription and mediates post-translational BMPR-II cleavage via the sheddases, ADAM10 and ADAM17 in pulmonary artery smooth muscle cells (PASMCs). TNFα-mediated suppression of BMPR-II subverts BMP signalling, leading to BMP6-mediated PASMC proliferation via preferential activation of an ALK2/ACTR-IIA signalling axis. Furthermore, TNFα, via SRC family kinases, increases pro-proliferative NOTCH2 signalling in HPAH PASMCs with reduced BMPR-II expression. We confirm this signalling switch in rodent models of PAH and demonstrate that anti-TNFα immunotherapy reverses disease progression, restoring normal BMP/NOTCH signalling. Collectively, these findings identify mechanisms by which BMP and TNFα signalling contribute to disease, and suggest a tractable approach for therapeutic intervention in PAH. Reduced BMP receptor II signalling underlies pulmonary arterial hypertension (PAH). Here, Hurst et al. show that TNFα subverts BMP signalling by increasing BMP6 expression and signalling via an alternative BMP receptor, ALK2, in pulmonary artery smooth muscle cells to drive abnormal proliferation and PAH.
Collapse
|
25
|
Vignozzi L, Morelli A, Cellai I, Filippi S, Comeglio P, Sarchielli E, Maneschi E, Vannelli GB, Adorini L, Maggi M. Cardiopulmonary protective effects of the selective FXR agonist obeticholic acid in the rat model of monocrotaline-induced pulmonary hypertension. J Steroid Biochem Mol Biol 2017; 165:277-292. [PMID: 27425465 DOI: 10.1016/j.jsbmb.2016.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 07/06/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
Abstract
Farnesoid X receptor (FXR) activation by obeticholic acid (OCA) has been demonstrated to inhibit inflammation and fibrosis development and even induce fibrosis regression in liver, kidney and intestine in multiple disease models. OCA also inhibits liver fibrosis in nonalcoholic steatohepatitis patients. FXR activation has also been demonstrated to suppress the inflammatory response and to promote lung repair after lung injury. This study investigated the effects of OCA treatment (3, 10 or 30mg/kg, daily for 5days a week, for 7 and/or 28 days) on inflammation, tissue remodeling and fibrosis in the monocrotaline (MCT)-induced pulmonary arterial hypertension (PAH) rat model. Treatment with OCA attenuated MCT-induced increased pulmonary arterial wall thickness and right ventricular hypertrophy, by i) blunting pathogenic inflammatory mechanisms (downregulation of interleukin 6, IL-6, and monocyte chemoattractant protein-1, MCP-1) and ii) enhancing protective mechanisms counteracting fibrosis and endothelial/mesenchymal transition. MCT-injected rats also showed a marked decrease of pulmonary artery responsiveness to both endothelium-dependent and independent relaxant stimuli, such as acetylcholine and a nitric oxide donor, sodium nitroprusside. Administration of OCA (30mg/kg) normalized this decreased responsiveness. Accordingly, OCA treatment induced profound beneficial effects on lung histology. In particular, both OCA doses markedly reduced the MCT-induced medial wall thickness increase in small pulmonary arteries. To evaluate the objective functional improvement by OCA treatment of MCT-induced PAH, we performed a treadmill test and measured duration of exercise. MCT significantly reduced, and OCA normalized treadmill endurance. Results with OCA were similar, or even superior, to those obtained with tadalafil, a well-established treatment of PAH. In conclusion, OCA treatment demonstrates cardiopulmonary protective effects, modulating lung vascular remodeling, reducing right ventricular hypertrophy and significantly improving exercise capacity. Thus, OCA can restore the balance between relaxant and contractile pathways in the lung, promoting cardiopulmonary protective actions.
Collapse
Affiliation(s)
- Linda Vignozzi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Annamaria Morelli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Ilaria Cellai
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Sandra Filippi
- Interdepartmental Laboratory of Functional and Cellular Pharmacology of Reproduction, Department of Neuroscience, Drug Research and Child Care, University of Florence, Florence, Italy
| | - Paolo Comeglio
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Erica Sarchielli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Elena Maneschi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | | | | | - Mario Maggi
- Sexual Medicine and Andrology Unit, Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy; I.N.B.B. (Istituto Nazionale Biostrutture e Biosistemi), Rome, Italy.
| |
Collapse
|
26
|
Guignabert C, Bailly S, Humbert M. Restoring BMPRII functions in pulmonary arterial hypertension: opportunities, challenges and limitations. Expert Opin Ther Targets 2016; 21:181-190. [DOI: 10.1080/14728222.2017.1275567] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Christophe Guignabert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Univ. Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Sabine Bailly
- INSERM U1036, Grenoble, France
- Laboratoire Biologie du Cancer et de l’Infection, Commissariat à l’Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, Grenoble, France
- Université Grenoble-Alpes, Grenoble, France
| | - Marc Humbert
- INSERM UMR_S 999, Le Plessis-Robinson, France
- Univ. Paris-Sud, Université Paris-Saclay, Kremlin-Bicêtre, France
- AP-HP, Service de Pneumologie, Centre de Référence de l’Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital de Bicêtre, France
| |
Collapse
|
27
|
BMPRII influences the response of pulmonary microvascular endothelial cells to inflammatory mediators. Pflugers Arch 2016; 468:1969-1983. [PMID: 27816994 DOI: 10.1007/s00424-016-1899-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/13/2016] [Accepted: 10/24/2016] [Indexed: 10/20/2022]
Abstract
Mutations in the bone morphogenetic protein receptor (BMPR2) gene have been observed in 70 % of patients with heritable pulmonary arterial hypertension (HPAH) and in 11-40 % with idiopathic PAH (IPAH). However, carriers of a BMPR2 mutation have only 20 % risk of developing PAH. Since inflammatory mediators are increased and predict survival in PAH, they could act as a second hit inducing the development of pulmonary hypertension in BMPR2 mutation carriers. Our specific aim was to determine whether inflammatory mediators could contribute to pulmonary vascular cell dysfunction in PAH patients with and without a BMPR2 mutation. Pulmonary microvascular endothelial cells (PMEC) and arterial smooth muscle cells (PASMC) were isolated from lung parenchyma of transplanted PAH patients, carriers of a BMPR2 mutation or not, and from lobectomy patients or lung donors. The effects of CRP and TNFα on mitogenic activity, adhesiveness capacity, and expression of adhesion molecules were investigated in PMECs and PASMCs. PMECs from BMPR2 mutation carriers induced an increase in PASMC mitogenic activity; moreover, endothelin-1 secretion by PMECs from carriers was higher than by PMECs from non-carriers. Recruitment of monocytes by PMECs isolated from carriers was higher compared to PMECs from non-carriers and from controls, with an elevated ICAM-1 expression. CRP increased adhesion of monocytes to PMECs in carriers and non-carriers, and TNFα only in carriers. PMEC from BMPR2 mutation carriers have enhanced adhesiveness for monocytes in response to inflammatory mediators, suggesting that BMPR2 mutation could generate susceptibility to an inflammatory insult in PAH.
Collapse
|
28
|
Huertas A, Phan C, Bordenave J, Tu L, Thuillet R, Le Hiress M, Avouac J, Tamura Y, Allanore Y, Jovan R, Sitbon O, Guignabert C, Humbert M. Regulatory T Cell Dysfunction in Idiopathic, Heritable and Connective Tissue-Associated Pulmonary Arterial Hypertension. Chest 2016; 149:1482-93. [DOI: 10.1016/j.chest.2016.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/27/2015] [Accepted: 01/04/2016] [Indexed: 12/21/2022] Open
|
29
|
Maruyama H, Dewachter C, Sakai S, Belhaj A, Rondelet B, Remmelink M, Vachiéry JL, Naeije R, Dewachter L. Bosentan reverses the hypoxia-induced downregulation of the bone morphogenetic protein signaling in pulmonary artery smooth muscle cells. Life Sci 2016; 159:111-115. [PMID: 27188586 DOI: 10.1016/j.lfs.2016.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 12/15/2022]
Abstract
AIMS Pulmonary hypertension (PH) is a common complication of chronic hypoxic lung diseases. Bone morphogenetic protein (BMP) and endothelin-1 signaling pathways have been shown to be altered in hypoxic PH and to play crucial roles in the associated pulmonary artery remodeling. We, therefore, aimed to study the potential link between hypoxia and the alteration of BMP and endothelin-1 signaling observed in pulmonary artery smooth muscle cells (PA-SMCs) in hypoxic PH. MATERIALS AND METHODS Human PA-SMCs were treated with hypoxia-mimetic agent cobalt chloride (CoCl2; 100μM), with or without pretreatment with a dual endothelin receptor antagonist bosentan (10μM). Expressions of preproendothelin-1 (PPET1), BMP type 2 receptor (BMPR-2), and one BMP signaling target gene, the inhibitor of DNA binding 1 (ID1) were evaluated by real time quantitative polymerase chain reaction. BMP2-treated PA-SMCs were assessed for Smad1/5/8 signaling activation by Western Blotting. KEY FINDINGS Treatment of PA-SMCs with CoCl2 increased PPET1 gene expression, while it did not alter expressions of endothelin converting enzyme, endothelin receptor type A or type B. Hypoxia-mimetic agent CoCl2 decreased the expressions of BMPR-2 and ID1 maximally after 3- and 6-hour treatment respectively, while CoCl2 treatment progressively increased noggin expression. Bosentan pretreatment restored expressions of BMPR-2 and ID1, as well as the activation (by phosphorylation) of Smad1/5/8 signaling induced by BMP2. SIGNIFICANCE Hypoxia induces the downregulation of the BMP signaling in PA-SMCs, at least, partly through the endothelin system. In hypoxic PH, increased endothelin-1 production might therefore contribute to the altered BMP signaling and subsequent PA-SMC hyperplasia.
Collapse
Affiliation(s)
- Hidekazu Maruyama
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium,.
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Satoshi Sakai
- Division of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Asmae Belhaj
- Department of Thoracic Surgery, Erasmus University Hospital, Brussels, Belgium
| | - Benoit Rondelet
- Department of Thoracic Surgery, Erasmus University Hospital, Brussels, Belgium
| | - Myriam Remmelink
- Department of Anatomopathology, Erasmus University Hospital, Brussels, Belgium
| | - Jean-Luc Vachiéry
- Department of Cardiology, Erasmus University Hospital, Brussels, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
30
|
Williams K, Andrie K, Cartoceti A, French S, Goldsmith D, Jennings S, Priestnall SL, Wilson D, Jutkowitz A. Pulmonary Veno-Occlusive Disease. Vet Pathol 2016; 53:813-22. [DOI: 10.1177/0300985815626572] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pulmonary hypertension is a well-known though poorly characterized disease in veterinary medicine. In humans, pulmonary veno-occlusive disease (PVOD) is a rare cause of severe pulmonary hypertension with a mean survival time of 2 years without lung transplantation. Eleven adult dogs (5 males, 6 females; median age 10.5 years, representing various breeds) were examined following the development of severe respiratory signs. Lungs of affected animals were evaluated morphologically and with immunohistochemistry for alpha smooth muscle actin, desmin, CD31, CD3, CD20, and CD204. All dogs had pulmonary lesions consistent with PVOD, consisting of occlusive remodeling of small- to medium-sized pulmonary veins, foci of pulmonary capillary hemangiomatosis (PCH), and accumulation of hemosiderophages; 6 of 11 dogs had substantial pulmonary arterial medial and intimal thickening. Ultrastructural examination and immunohistochemistry showed that smooth muscle cells contributed to the venous occlusion. Increased expression of CD31 was evident in regions of PCH indicating increased numbers of endothelial cells in these foci. Spindle cells strongly expressing alpha smooth muscle actin and desmin co-localized with foci of PCH; similar cells were present but less intensely labeled elsewhere in non-PCH alveoli. B cells and macrophages, detected by immunohistochemistry, were not co-localized with the venous lesions of canine PVOD; small numbers of CD3-positive T cells were occasionally in and around the wall of remodeled veins. These findings indicate a condition in dogs with clinically severe respiratory disease and pathologic features resembling human PVOD, including foci of pulmonary venous remodeling and PCH.
Collapse
Affiliation(s)
- K. Williams
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - K. Andrie
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - A. Cartoceti
- School of Veterinary Medicine, University of California, Davis, CA, USA
| | - S. French
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| | - D. Goldsmith
- School of Veterinary Medicine, University of California, Davis, CA, USA
| | - S. Jennings
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA, USA
| | | | - D. Wilson
- School of Veterinary Medicine, University of California, Davis, CA, USA
| | - A. Jutkowitz
- College of Veterinary Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
31
|
Sahraoui A, Dewachter C, de Medina G, Naeije R, Aouichat Bouguerra S, Dewachter L. Myocardial Structural and Biological Anomalies Induced by High Fat Diet in Psammomys obesus Gerbils. PLoS One 2016; 11:e0148117. [PMID: 26840416 PMCID: PMC4740502 DOI: 10.1371/journal.pone.0148117] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 01/13/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Psammomys obesus gerbils are particularly prone to develop diabetes and obesity after brief period of abundant food intake. A hypercaloric high fat diet has been shown to affect cardiac function. Here, we sought to determine whether a short period of high fat feeding might alter myocardial structure and expression of calcium handling proteins in this particular strain of gerbils. METHODS Twenty Psammomys obesus gerbils were randomly assigned to receive a normal plant diet (controls) or a high fat diet. At baseline and 16-week later, body weight, plasma biochemical parameters (including lipid and carbohydrate levels) were evaluated. Myocardial samples were collected for pathobiological evaluation. RESULTS Sixteen-week high fat dieting resulted in body weight gain and hyperlipidemia, while levels of carbohydrates remained unchanged. At myocardial level, high fat diet induced structural disorganization, including cardiomyocyte hypertrophy, lipid accumulation, interstitial and perivascular fibrosis and increased number of infiltrating neutrophils. Myocardial expressions of pro-apoptotic Bax-to-Bcl-2 ratio, pro-inflammatory cytokines [interleukin (IL)-1β and tumor necrosis factor (TNF)-α], intercellular (ICAM1) and vascular adhesion molecules (VCAM1) increased, while gene encoding cardiac muscle protein, the alpha myosin heavy polypeptide (MYH6), was downregulated. Myocardial expressions of sarco(endo)plasmic calcium-ATPase (SERCA2) and voltage-dependent calcium channel (Cacna1c) decreased, while protein kinase A (PKA) and calcium-calmodulin-dependent protein kinase (CaMK2D) expressions increased. Myocardial expressions of ryanodine receptor, phospholamban and sodium/calcium exchanger (Slc8a1) did not change. CONCLUSIONS We conclude that a relative short period of high fat diet in Psammomys obesus results in severe alterations of cardiac structure, activation of inflammatory and apoptotic processes, and altered expression of calcium-cycling determinants.
Collapse
Affiliation(s)
- Abdelhamid Sahraoui
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
- Team of Cellular and Molecular Physiopathology, Faculty of Biological Sciences, Houari Boumediene University of Sciences and Technology, El Alia, Algiers, Algeria
| | - Céline Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Geoffrey de Medina
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Robert Naeije
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Souhila Aouichat Bouguerra
- Team of Cellular and Molecular Physiopathology, Faculty of Biological Sciences, Houari Boumediene University of Sciences and Technology, El Alia, Algiers, Algeria
| | - Laurence Dewachter
- Laboratory of Physiology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
32
|
Abstract
Previously considered a disease isolated to the pulmonary circulation, pulmonary arterial hypertension is now being recognized as a systemic disorder that is associated with significant metabolic dysfunction. Numerous animal models have demonstrated the development of pulmonary arterial hypertension following the onset of insulin resistance, indicating that insulin resistance may be causal. Recent publications highlighting alterations in aerobic glycolysis, fatty acid oxidation, and the tricarboxylic acid cycle in the pulmonary circulation and right ventricle have expanded our understanding of the complex pathobiology of this disease. By targeting these derangements in metabolism, numerous researchers are investigating noninvasive techniques to monitor disease activity and therapeutics that address the underlying metabolic condition. In the following review, we will explore pre-clinical and clinical studies investigating the metabolic dysfunction seen in pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Tufik R Assad
- Division of Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, T1218 Medical Center North, 1161 21st Avenue South, Nashville, TN, 37232, USA
| | | |
Collapse
|
33
|
Zeng Y, Liu H, Kang K, Wang Z, Hui G, Zhang X, Zhong J, Peng W, Ramchandran R, Raj JU, Gou D. Hypoxia inducible factor-1 mediates expression of miR-322: potential role in proliferation and migration of pulmonary arterial smooth muscle cells. Sci Rep 2015; 5:12098. [PMID: 26166214 PMCID: PMC4499844 DOI: 10.1038/srep12098] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 06/15/2015] [Indexed: 01/08/2023] Open
Abstract
There is growing evidence that microRNAs play important roles in cellular responses to hypoxia and in pulmonary hypertensive vascular remodeling, but the exact molecular mechanisms involved are not fully elucidated. In this study, we identified miR-322 as one of the microRNAs induced in lungs of chronically hypoxic mice and rats. The expression of miR-322 was also upregulated in primary cultured rat pulmonary arterial smooth muscle cells (PASMC) in response to hypoxia. We demonstrated that HIF-1α, but not HIF-2α, transcriptionally upregulates the expression of miR-322 in hypoxia. Furthermore, miR-322 facilitated the accumulation of HIF-1α in the nucleus and promoted hypoxia-induced cell proliferation and migration. Direct targeting BMPR1a and smad5 by miR-322 was demonstrated in PASMCs suggesting that downregulation of BMP-Smad signaling pathway may be mediating the hypoxia-induced PASMC proliferation and migration. Our study implicates miR-322 in the hypoxic proliferative response of PASMCs suggesting that it may be playing a role in pulmonary vascular remodeling associated with pulmonary hypertension.
Collapse
Affiliation(s)
- Yan Zeng
- 1] Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresourse and Eco-environmental Science, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China [2] Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Hongtao Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresourse and Eco-environmental Science, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Kang Kang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresourse and Eco-environmental Science, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Zhiwei Wang
- Department of Cardiovascular Surgery, Shenzhen Sun Yat-Sen Cardiovascular Hospital, Shenzhen, Guangdong, 518000, China
| | - Gang Hui
- Department of Chest Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518000, China
| | - Xiaoying Zhang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresourse and Eco-environmental Science, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Jiasheng Zhong
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresourse and Eco-environmental Science, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Wenda Peng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, 518060, China
| | - Ramaswamy Ramchandran
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| | - J Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| | - Deming Gou
- 1] Shenzhen Key Laboratory of Microbial Genetic Engineering, Shenzhen Key Laboratory of Marine Bioresourse and Eco-environmental Science, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong, 518060, China [2] Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, U.S.A
| |
Collapse
|
34
|
Altered pulmonary artery endothelial-smooth muscle cell interactions in experimental congenital diaphragmatic hernia. Pediatr Res 2015; 77:511-9. [PMID: 25580737 PMCID: PMC4363155 DOI: 10.1038/pr.2015.13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 10/19/2014] [Indexed: 11/24/2022]
Abstract
BACKGROUND Pulmonary hypertension (PH) secondary to vascular remodeling contributes to poor outcomes in congenital diaphragmatic hernia (CDH), however mechanisms responsible are unknown. We hypothesized that pulmonary artery endothelial cell (PAEC) dysfunction contributes to smooth muscle cell (SMC) hyperplasia in experimental CDH. METHODS PAEC and SMC were isolated from fetal sheep with experimental CDH and controls. SMC growth was assessed alone and with SOD plus catalase and during coculture with control or CDH PAEC with and without ET-1 siRNA transfection. ET-1 protein was measured in PAEC and SMC lysates and supernatant. ROS production was measured in normal and CDH PAECs with and without ET-1 siRNA. PAEC growth and tube formation were measured with SOD plus catalase. RESULTS CDH SMC growth was decreased and increased with coculture with CDH PAEC more than control PAEC. Treatment of CDH PAEC with SOD plus catalase or ET-1 siRNA prevented the increase in SMC growth seen with coculture. ET-1 protein was increased in CDH PAEC and SMC. ROS production was increased in CDH PAEC and decreased with ET-1 SiRNA. SOD plus catalase restored CDH PAEC growth and tube formation. CONCLUSION PAEC dysfunction in experimental CDH increases SMC proliferation via ET-1 induced ROS production by PAEC.
Collapse
|
35
|
Endothelin-Bone morphogenetic protein type 2 receptor interaction induces pulmonary artery smooth muscle cell hyperplasia in pulmonary arterial hypertension. J Heart Lung Transplant 2015; 34:468-78. [DOI: 10.1016/j.healun.2014.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 08/08/2014] [Accepted: 09/18/2014] [Indexed: 10/24/2022] Open
|
36
|
Zeng Y, Pan Y, Liu H, Kang K, Wu Y, Hui G, Peng W, Ramchandran R, Raj JU, Gou D. MiR-20a regulates the PRKG1 gene by targeting its coding region in pulmonary arterial smooth muscle cells. FEBS Lett 2014; 588:4677-85. [PMID: 25447536 DOI: 10.1016/j.febslet.2014.10.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 10/22/2014] [Accepted: 10/29/2014] [Indexed: 01/07/2023]
Abstract
Chronic hypoxia triggers pulmonary vascular remodeling, which is associated with de-differentiation of pulmonary artery smooth muscle cells (PASMC). Here, we show that miR-20a expression is up-regulated in response to hypoxia in both mouse and human PASMC. We also observed that miR-20a represses the protein kinase, cGMP-dependent, type I (PRKG1) gene and we identified two crucial miR-20a binding sites within the coding region of PRKG1. Functional studies showed that miR-20a promotes the proliferation and migration of human PASMC, whereas it inhibits their differentiation. In summary, we provided a possible mechanism by which hypoxia results in decreased PRKG1 expression and in the phenotypic switching of PASMC.
Collapse
Affiliation(s)
- Yan Zeng
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Yanping Pan
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Hongtai Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Kang Kang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Yike Wu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Gang Hui
- Department of Chest Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518000, China
| | - Wenda Peng
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | | | - J Usha Raj
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences, Shenzhen University, Shenzhen, Guangdong 518060, China; Department of Pediatrics, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
37
|
Jiang Q, Fu X, Tian L, Chen Y, Yang K, Chen X, Zhang J, Lu W, Wang J. NOX4 mediates BMP4-induced upregulation of TRPC1 and 6 protein expressions in distal pulmonary arterial smooth muscle cells. PLoS One 2014; 9:e107135. [PMID: 25203114 PMCID: PMC4159322 DOI: 10.1371/journal.pone.0107135] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/10/2014] [Indexed: 02/07/2023] Open
Abstract
Rationale Our previous studies demonstrated that bone morphogenetic protein 4 (BMP4) mediated, elevated expression of canonical transient receptor potential (TRPC) largely accounts for the enhanced proliferation in pulmonary arterial smooth muscle cells (PASMCs). In the present study, we sought to determine the signaling pathway through which BMP4 up-regulates TRPC expression. Methods We employed recombinant human BMP4 (rhBMP4) to determine the effects of BMP4 on NADPH oxidase 4 (NOX4) and reactive oxygen species (ROS) production in rat distal PASMCs. We also designed small interfering RNA targeting NOX4 (siNOX4) and detected whether NOX4 knockdown affects rhBMP4-induced ROS, TRPC1 and 6 expression, cell proliferation and intracellular Ca2+ determination in PASMCs. Results In rhBMP4 treated rat distal PASMCs, NOX4 expression was (226.73±11.13) %, and the mean ROS level was (123.65±1.62) % of that in untreated control cell. siNOX4 transfection significantly reduced rhBMP4-induced elevation of the mean ROS level in PASMCs. Moreover, siNOX4 transfection markedly reduced rhBMP4-induced elevation of TRPC1 and 6 proteins, basal [Ca2+]i and SOCE. Furthermore, compared with control group (0.21±0.001), the proliferation of rhBMP4 treated cells was significantly enhanced (0.41±0.001) (P<0.01). However, such increase was attenuated by knockdown of NOX4. Moreover, external ROS (H2O2 100 µM, 24 h) rescued the effects of NOX4 knockdown, which included the declining of TRPC1 and 6 expression, basal intracellular calcium concentration ([Ca2+]i) and store-operated calcium entry (SOCE), suggesting that NOX4 plays as an important mediator in BMP4-induced proliferation and intracellular calcium homeostasis. Conclusion These results suggest that BMP4 may increase ROS level, enhance TRPC1 and 6 expression and proliferation by up-regulating NOX4 expression in PASMCs.
Collapse
Affiliation(s)
- Qian Jiang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xin Fu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lichun Tian
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kai Yang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Xiuqing Chen
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jie Zhang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- * E-mail: (JW); (WL)
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, The 1st Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Pulmonary, Inner Mongolia People’s Hospital, Huhhot, Inner Mongolia, China
- * E-mail: (JW); (WL)
| |
Collapse
|
38
|
Wang RN, Green J, Wang Z, Deng Y, Qiao M, Peabody M, Zhang Q, Ye J, Yan Z, Denduluri S, Idowu O, Li M, Shen C, Hu A, Haydon RC, Kang R, Mok J, Lee MJ, Luu HL, Shi LL. Bone Morphogenetic Protein (BMP) signaling in development and human diseases. Genes Dis 2014; 1:87-105. [PMID: 25401122 PMCID: PMC4232216 DOI: 10.1016/j.gendis.2014.07.005] [Citation(s) in RCA: 712] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 07/15/2014] [Indexed: 02/06/2023] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are a group of signaling molecules that belongs to the Transforming Growth Factor-β (TGF-β) superfamily of proteins. Initially discovered for their ability to induce bone formation, BMPs are now known to play crucial roles in all organ systems. BMPs are important in embryogenesis and development, and also in maintenance of adult tissue homeostasis. Mouse knockout models of various components of the BMP signaling pathway result in embryonic lethality or marked defects, highlighting the essential functions of BMPs. In this review, we first outline the basic aspects of BMP signaling and then focus on genetically manipulated mouse knockout models that have helped elucidate the role of BMPs in development. A significant portion of this review is devoted to the prominent human pathologies associated with dysregulated BMP signaling.
Collapse
Affiliation(s)
- Richard N. Wang
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jordan Green
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zhongliang Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Youlin Deng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Min Qiao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Michael Peabody
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Qian Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Jixing Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Bioengineering, Chongqing University, Chongqing, China
| | - Zhengjian Yan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery, Medicine, and Gynecology, the Affiliated Hospitals of Chongqing Medical University, Chongqing 400016, China
| | - Sahitya Denduluri
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Olumuyiwa Idowu
- The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Melissa Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Christine Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Alan Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Richard Kang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - James Mok
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue L. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
39
|
Feng J, Gao J, Li Y, Yang Y, Dang L, Ye Y, Deng J, Li A. BMP4 enhances foam cell formation by BMPR-2/Smad1/5/8 signaling. Int J Mol Sci 2014; 15:5536-52. [PMID: 24690996 PMCID: PMC4013580 DOI: 10.3390/ijms15045536] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/09/2014] [Accepted: 02/12/2014] [Indexed: 01/10/2023] Open
Abstract
Atherosclerosis and its complications are characterized by lipid-laden foam cell formation. Recently, an obvious up-regulation of BMP4 was observed in atherosclerotic plaque, however, its function and the underlying mechanism remains unknown. In our study, BMP4 pretreatment induced macrophage foam cell formation. Furthermore, a dramatic increase in the ratio of cholesteryl ester (CE) to total cholesterol (TC) was observed in BMP4-treated macrophages, accompanied by the reduction of cholesterol outflow. Importantly, BMP4 stimulation inhibited the expression levels of the two most important cellular cholesterol transporters ABCA1 and ABCG1, indicating that BMP4 may induce formation of foam cells by attenuating transporters expression. Further mechanism analysis showed that BMPR-2, one of the BMP4 receptors, was significantly increased in BMP4 treated macrophage foam cells. That blocking its expression using specific siRNA significantly increased ABCA1 and ABCG1 levels. Additionally, BMP4 treatment triggered the activation of Smad1/5/8 pathway by BMPR-2 signaling. After blocking the Smad1/5/8 with its inhibitor, ABCA1 and ABCG1 expression levels were up-regulated significantly, suggesting that BMP4 inhibited the expression of ABCA1 and ABCG1 through the BMPR-2/Smad1/2/8 signaling pathway. Therefore, our results will provide a new insight about how BMP4 accelerate the progressio of atherosclerosis, and it may become a potential target against atherosclerosis and its complications.
Collapse
Affiliation(s)
- Jun Feng
- Department of Cerebral Vessels, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Jiangfei Gao
- Department of Neurology, Shangluo Central Hospital, Shangluo 726000, Shaanxi, China.
| | - Yuxin Li
- Department of Neurology, the Second Affiliated Hospital, Xi'an Medical College, Xi'an 710038, Shaanxi, China.
| | - Yanhua Yang
- Department of Neurology, Shaanxi Armed Police Corps Hospital, Xi'an 710054, Shaanxi, China.
| | - Lili Dang
- Department of Neurology, Xingyuan Hospital, Yulin 719000, Shaanxi, China.
| | - Yuanpeng Ye
- Department of Cerebral Vessels, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Jingyuan Deng
- Department of Rehabilitation Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Antai Li
- Department of Neurology, Xi'an Central Hospital, Xi'an 710003, Shaanxi, China.
| |
Collapse
|
40
|
Gien J, Tseng N, Seedorf G, Roe G, Abman SH. Peroxisome proliferator activated receptor-γ-Rho-kinase interactions contribute to vascular remodeling after chronic intrauterine pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2013; 306:L299-308. [PMID: 24375792 DOI: 10.1152/ajplung.00271.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) and Rho-kinase (ROCK) regulate smooth muscle cell (SMC) proliferation and contribute to vascular remodeling in adult pulmonary hypertension. Whether these pathways interact to contribute to the development of vascular remodeling in persistent pulmonary hypertension of the newborn (PPHN) remains unknown. We hypothesized that ROCK-PPARγ interactions increase SMC proliferation resulting in vascular remodeling in experimental PPHN. Pulmonary artery SMCs (PASMCs) were harvested from fetal sheep after partial ligation of the ductus arteriosus in utero (PPHN) and controls. Cell counts were performed daily for 5 days with or without PPARγ agonists and ROCK inhibition. PPARγ and ROCK protein expression/activity were measured by Western blot in normal and PPHN PASMCs. We assessed PPARγ-ROCK interactions by studying the effect of ROCK activation on PPARγ activity and PPARγ inhibition (siRNA) on ROCK activity and PASMC proliferation. At baseline, PPHN PASMC cell number was increased by 38% above controls on day 5. ROCK protein expression/activity were increased by 25 and 34% and PPARγ protein/activity decreased by 40 and 50% in PPHN PASMC. ROCK inhibition and PPARγ activation restored PPHN PASMC growth to normal values. ROCK inhibition increased PPARγ activity by 50% in PPHN PASMC, restoring PPARγ activity to normal. In normal PASMCs, ROCK activation decreased PPARγ activity and PPARγ inhibition increased ROCK activity and cell proliferation, resulting in a PPHN hyperproliferative PASMC phenotype. PPARγ-ROCK interactions regulate SMC proliferation and contribute to increased PPHN PASMC proliferation and vascular remodeling in PPHN. Restoring normal PPARγ-ROCK signaling may prevent vascular remodeling and improve outcomes in PPHN.
Collapse
Affiliation(s)
- Jason Gien
- Perinatal Research Facility, 13243 E. 23rd Ave., Mail Stop F441, Aurora, CO 80045.
| | | | | | | | | |
Collapse
|
41
|
Renteria LS, Austin M, Lazaro M, Andrews MA, Lustina J, Raj JU, Ibe BO. RhoA-Rho kinase and platelet-activating factor stimulation of ovine foetal pulmonary vascular smooth muscle cell proliferation. Cell Prolif 2013; 46:563-75. [PMID: 24033386 PMCID: PMC3788060 DOI: 10.1111/cpr.12052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 05/17/2013] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Platelet-activating factor (PAF) is produced by pulmonary vascular smooth muscle cells (PVSMC). We studied effects of Rho kinase on PAF stimulation of PVSMC proliferation in an attempt to understand the role of RhoA/Rho kinase on PAF-induced ovine foetal pulmonary vascular remodelling. Our hypothesis is that PAF acts through Rho kinase, as one of its downstream signals, to induce arterial (SMC-PA) and venous (SMC-PV) cell proliferation in the hypoxic lung environment of the foetus, in utero. MATERIALS AND METHODS Rho kinase and MAPK effects on PAF receptor (PAFR)-mediated cell population expansion, and PAFR expression, were studied by DNA synthesis, western blot analysis and immunocytochemistry. Effects of constructs T19N and G14V on PAF-induced cell proliferation were also investigated. RESULTS Hypoxia increased PVSMC proliferation and Rho kinase inhibitors, Y-27632 and Fasudil (HA-1077) as well as MAPK inhibitors PD 98059 and SB 203580 attenuated PAF stimulation of cell proliferation. RhoA T19N and G14V stimulated cell proliferation, but co-incubation with PAF did not affect proliferative effects of the constructs. PAFR protein expression was significantly downregulated in both cell types by both Y-27632 and HA-1077, with comparable profiles. Also, cells treated with Y-27632 had less PAF receptor fluorescence with significant disruption of cell morphology. CONCLUSIONS Our results show that Rho kinase non-specifically modulated PAFR-mediated responses by a translational modification of PAFR protein, and suggest that, in vivo, activation of Rho kinase by PAF may be a further pathway to sustain PAFR-mediated PVSMC proliferation.
Collapse
Affiliation(s)
- L. S. Renteria
- Division of NeonatologyDepartment of PediatricsLos Angeles Biomedical Research Institute at Harbor‐UCLA Medical CenterTorranceCA90502USA
| | - M. Austin
- Division of NeonatologyDepartment of PediatricsLos Angeles Biomedical Research Institute at Harbor‐UCLA Medical CenterTorranceCA90502USA
| | - M. Lazaro
- Division of NeonatologyDepartment of PediatricsLos Angeles Biomedical Research Institute at Harbor‐UCLA Medical CenterTorranceCA90502USA
| | - M. A. Andrews
- Division of NeonatologyDepartment of PediatricsLos Angeles Biomedical Research Institute at Harbor‐UCLA Medical CenterTorranceCA90502USA
| | - J. Lustina
- Division of NeonatologyDepartment of PediatricsLos Angeles Biomedical Research Institute at Harbor‐UCLA Medical CenterTorranceCA90502USA
| | - J. U. Raj
- Department of PediatricsUniversity of Illinois ChicagoChicagoIL60612USA
| | - B. O. Ibe
- Division of NeonatologyDepartment of PediatricsLos Angeles Biomedical Research Institute at Harbor‐UCLA Medical CenterTorranceCA90502USA
| |
Collapse
|
42
|
Li X, Lu W, Fu X, Zhang Y, Yang K, Zhong N, Ran P, Wang J. BMP4 increases canonical transient receptor potential protein expression by activating p38 MAPK and ERK1/2 signaling pathways in pulmonary arterial smooth muscle cells. Am J Respir Cell Mol Biol 2013; 49:212-20. [PMID: 23526217 DOI: 10.1165/rcmb.2012-0051oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Abnormal bone morphogenetic protein (BMP) signaling has been implicated in the pathogenesis of pulmonary hypertension. We previously found that BMP4 elevated basal intracellular Ca(2+) ([Ca(2+)]i) concentrations in distal pulmonary arterial smooth muscle cells (PASMCs), attributable in large part to enhanced store-operated Ca(2+) entry through store-operated Ca(2+) channels (SOCCs). Moreover, BMP4 up-regulated the expression of canonical transient receptor potential (TRPC) proteins thought to compose SOCCs. The present study investigated the signaling pathways through which BMP4 regulates TRPC expression and basal [Ca(2+)]i in distal PASMCs. Real-time quantitative PCR was used for the measurement of mRNA, Western blotting was used for the measurement of protein, and fluorescent microscopic for [Ca(2+)]i was used to determine the involvement of p38 and extracellular regulated kinase (ERK)-1/2 mitogen-activated protein kinase (MAPK) signaling in BMP4-induced TRPC expression and the elevation of [Ca(2+)]i in PASMCs. We found that the treatment of BMP4 led to the activation of both p38 MAPK and ERK1/2 in rat distal PASMCs. The induction of TRPC1, TRPC4, and TRPC6 expression, and the increases of [Ca(2+)]i caused by BMP4 in distal PASMCs, were inhibited by treatment with either SB203580 (10 μM), the selective inhibitor for p38 activation, or the specific p38 small interfering RNA (siRNA). Similarly, those responses induced by BMP4 were also abolished by treatment with PD98059 (5 μM), the selective inhibitor of ERK1/2, or by the knockdown of ERK1/2 using its specific siRNA. These results indicate that BMP4 participates in the regulation of Ca(2+) signaling in PASMCs by modulating TRPC channel expression via activating p38 and ERK1/2 MAPK pathways.
Collapse
Affiliation(s)
- Xiaoyan Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Ogo T, Chowdhury HM, Yang J, Long L, Li X, Torres Cleuren YN, Morrell NW, Schermuly RT, Trembath RC, Nasim MT. Inhibition of overactive transforming growth factor-β signaling by prostacyclin analogs in pulmonary arterial hypertension. Am J Respir Cell Mol Biol 2013; 48:733-41. [PMID: 23418342 DOI: 10.1165/rcmb.2012-0049oc] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The heterozygous loss of function mutations in the Type II bone morphogenetic protein receptor (BMPR-II), a member of the transforming growth factor (TGF-β) receptor family, underlies the majority of familial cases of pulmonary arterial hypertension (PAH). The TGF-β1 pathway is activated in PAH, and inhibitors of TGF-β1 signaling prevent the development and progression of PAH in experimental models. However, the effects of currently used therapies on the TGF-β pathway remain unknown. Prostacyclin analogs comprise the first line of treatment for clinical PAH. We hypothesized that these agents effectively decrease the activity of the TGF-β1 pathway. Beraprost sodium (BPS), a prostacyclin analog, selectively inhibits proliferation in a dose-dependent manner in murine primary pulmonary arterial smooth muscle cells (PASMCs) harboring a pathogenic BMPR2 nonsense mutation in both the presence and absence of TGF-β1 stimulation. Our study demonstrates that this agent inhibits TGF-β1-induced SMAD-dependent and SMAD-independent signaling via a protein kinase A-dependent pathway by reducing the phosphorylation of SMADs 2 and 3 and p38 mitogen-activated protein kinase proteins. Finally, in a monocrotaline-induced rat model of PAH, which is associated with increased TGF-β signaling, this study confirms that treprostinil, a stable prostacyclin analog, inhibits the TGF-β pathway by reducing SMAD3 phosphorylation. Taken together, these data suggest that prostacyclin analogs inhibit dysregulated TGF-β signaling in vitro and in vivo, and reduce BMPR-II-mediated proliferation defects in mutant mice PASMCs.
Collapse
Affiliation(s)
- Takeshi Ogo
- Department of Medical and Molecular Genetics, School of Medicine, King's College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Makanga M, Dewachter C, Maruyama H, Vuckovic A, Rondelet B, Naeije R, Dewachter L. Downregulated bone morphogenetic protein signaling in nitrofen-induced congenital diaphragmatic hernia. Pediatr Surg Int 2013; 29:823-34. [PMID: 23832098 DOI: 10.1007/s00383-013-3340-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/24/2013] [Indexed: 12/14/2022]
Abstract
PURPOSE Bone morphogenetic proteins (BMP) have been shown to play crucial roles in not only lung and heart development, but also in the pathogenesis of pulmonary vascular remodeling in pulmonary hypertension (PH). We therefore hypothesized that BMP signaling could be altered in nitrofen-induced congenital diaphragmatic hernia (CDH) and associated PH. METHODS Pregnant rats were exposed to either 100 mg nitrofen or vehicle on embryonic day (E) 9.5. On E17 and E21, fetuses were delivered by cesarean section, killed and checked for left-sided CDH. The tissue was then harvested for pathobiological evaluation. RESULTS In nitrofen-induced CDH, pulmonary expressions of BMP4, BMP receptor (BMPR) type 2 and Id1 decreased on E17 and E21. On E17, pulmonary gremlin-1 expression increased, while BMP7 decreased. In the lungs, Id1 expression was correlated to BMP4 and BMPR2 and inversely correlated to gremlin-1 expression. Myocardial expressions of BMPR2, BMPR1A, BMP7 and SERCA-2A decreased, while gremlin-1 and noggin expressions increased on E17. On E21, myocardial expressions of Id1 and SERCA-2A decreased, while gremlin-1 expression increased. Moreover, BMPR2 and BMPR1A expressions were correlated to SERCA-2A expression and inversely correlated to pro-apoptotic Bax/Bcl2 ratio within the myocardium. CONCLUSION Downregulation of BMP signaling seems to contribute to pulmonary and myocardial anomalies observed in nitrofen-induced CDH.
Collapse
Affiliation(s)
- Martine Makanga
- Laboratory of Physiology and Physiopathology, Faculty of Medicine, Université Libre de Bruxelles, 808, Lennik Road, 1070, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
45
|
Kudryavtseva O, Aalkjaer C, Matchkov VV. Vascular smooth muscle cell phenotype is defined by Ca2+-dependent transcription factors. FEBS J 2013; 280:5488-99. [PMID: 23848563 DOI: 10.1111/febs.12414] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 06/21/2013] [Accepted: 06/26/2013] [Indexed: 12/12/2022]
Abstract
Ca(2+) is an important second messenger in vascular smooth muscle cells (VSMCs). Therefore, VSMCs exercise tight control of the intracellular Ca(2+) concentration ([Ca(2+)]i) by expressing a wide repertoire of Ca(2+) channels and transporters. The presence of several pathways for Ca(2+) influx and efflux provides many possibilities for controlling [Ca(2+)]i in a spatial and temporal manner. Intracellular Ca(2+) has a dual role in VSMCs; first, it is necessary for VSMC contraction; and, second, it can activate multiple transcription factors. These factors are cAMP response element-binding protein, nuclear factor of activated T lymphocytes, and serum response factor. Furthermore, it was recently reported that the C-terminus of voltage-dependent L-type Ca(2+) calcium channels can regulate transcription in VSMCs. Transcription regulation in VSMCs modulates the expression patterns of genes, including genes coding for contractile and cytoskeleton proteins, and those promoting proliferation and cell growth. Depending on their gene expression, VSMCs can exist in different functional states or phenotypes. The majority of healthy VSMCs show a contractile phenotype, characterized by high contractile ability and a low proliferative rate. However, VSMCs can undergo phenotypic modulation with different physiological and pathological stimuli, whereby they start to proliferate, migrate, and synthesize excessive extracellular matrix. These events are associated with injury repair and angiogenesis, but also with the development of cardiovascular pathologies, such as atherosclerosis and hypertension. This review discusses the currently known Ca(2+)-dependent transcription factors in VSMCs, their regulation by Ca(2+) signalling, and their role in the VSMC phenotype.
Collapse
|
46
|
Gosemann JH, Friedmacher F, Fujiwara N, Alvarez LAJ, Corcionivoschi N, Puri P. Disruption of the bone morphogenetic protein receptor 2 pathway in nitrofen-induced congenital diaphragmatic hernia. ACTA ACUST UNITED AC 2013; 98:304-9. [PMID: 23780850 DOI: 10.1002/bdrb.21065] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 04/19/2013] [Indexed: 11/07/2022]
Abstract
BACKGROUND/PURPOSE Congenital diaphragmatic hernia (CDH) remains a major therapeutic challenge despite advances in neonatal resuscitation and intensive care. The high mortality and morbidity in CDH has been attributed to pulmonary hypoplasia and persistent pulmonary hypertension (PH). Bone morphogenetic protein receptor 2 (BMPR2) plays a key role in pulmonary vasculogenesis during the late stages of fetal lung development. BMPR2 is essential for control of endothelial and smooth muscle cell proliferation. Dysfunction of BMPR2 and downstream signaling have been shown to disturb the crucial balance of proliferation of smooth muscle cells contributing to the pathogenesis of human and experimental PH. We designed this study to investigate the hypothesis that BMPR2 signaling is disrupted in nitrofen-induced CDH. METHODS Pregnant rats were treated with nitrofen or vehicle on gestational day 9 (D9). Fetuses were sacrificed on D21 and divided into CDH and control. Quantitative real-time polymerase chain reaction, Western blotting, and confocal-immunofluorescence were performed to determine pulmonary gene expression levels and protein expression of BMPR2 and related proteins. RESULTS Pulmonary Bmpr2 gene expression levels were significantly decreased in nitrofen-induced CDH compared to controls. Western blotting and confocal microscopy revealed decreased pulmonary BMPR2 protein expression and increased activation of p38(MAPK) in CDH compared to controls. CONCLUSION The observed disruption of the BMPR2 signaling pathway may lead to extensive vascular remodeling and contribute to PH in the nitrofen-induced CDH model. BMPR2 may therefore represent a potential target for the treatment of PH in CDH.
Collapse
Affiliation(s)
- Jan-Hendrik Gosemann
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Department of Pediatric Surgery, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
47
|
Dunmore BJ, Drake KM, Upton PD, Toshner MR, Aldred MA, Morrell NW. The lysosomal inhibitor, chloroquine, increases cell surface BMPR-II levels and restores BMP9 signalling in endothelial cells harbouring BMPR-II mutations. Hum Mol Genet 2013; 22:3667-79. [PMID: 23669347 PMCID: PMC3749859 DOI: 10.1093/hmg/ddt216] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by dysregulated pulmonary artery endothelial cell (PAEC) proliferation, apoptosis and permeability. Loss-of-function mutations in the bone morphogenetic protein receptor type-II (BMPR-II) are the most common cause of heritable PAH, usually resulting in haploinsufficiency. We previously showed that BMPR-II expression is regulated via a lysosomal degradative pathway. Here, we show that the antimalarial drug, chloroquine, markedly increased cell surface expression of BMPR-II protein independent of transcription in PAECs. Inhibition of protein synthesis experiments revealed a rapid turnover of cell surface BMPR-II, which was inhibited by chloroquine treatment. Chloroquine enhanced PAEC expression of BMPR-II following siRNA knockdown of the BMPR-II transcript. Using blood outgrowth endothelial cells (BOECs), we confirmed that signalling in response to the endothelial BMPR-II ligand, BMP9, is compromised in BOECs from patients harbouring BMPR-II mutations, and in BMPR-II mutant PAECs. Chloroquine significantly increased gene expression of BMP9-BMPR-II signalling targets Id1, miR21 and miR27a in both mutant BMPR-II PAECs and BOECs. These findings provide support for the restoration of cell surface BMPR-II with agents such as chloroquine as a potential therapeutic approach for heritable PAH.
Collapse
Affiliation(s)
- Benjamin J Dunmore
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
48
|
Long L, Yang X, Southwood M, Lu J, Marciniak SJ, Dunmore BJ, Morrell NW. Chloroquine prevents progression of experimental pulmonary hypertension via inhibition of autophagy and lysosomal bone morphogenetic protein type II receptor degradation. Circ Res 2013; 112:1159-70. [PMID: 23446737 DOI: 10.1161/circresaha.111.300483] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
RATIONALE Pulmonary arterial hypertension (PAH) is characterized by excessive proliferation and apoptosis resistance in pulmonary artery smooth muscle cells (PASMCs). OBJECTIVE We reasoned that chloroquine, based on its ability to inhibit autophagy and block lysosomal degradation of the bone morphogenetic protein type II receptor (BMPR-II), might exert beneficial effects in this disease. METHODS AND RESULTS PAH was induced in male Sprague-Dawley rats by administering monocrotaline. The induction of PAH was associated with changes in lung expression of LC3B-II, ATG5, and p62, consistent with increased autophagy, and decreased BMPR-II protein expression. Administration of chloroquine prevented the development of PAH, right ventricular hypertrophy, and vascular remodelling after monocrotaline, and prevented progression of established PAH in this model. Similar results were obtained with hydroxychloroquine. Chloroquine treatment increased whole lung and PASMC p62 protein levels consistent with inhibition of autophagy, and increased levels of BMPR-II protein. Chloroquine inhibited proliferation and increased apoptosis of PASMCs in vivo. In cultured rat PASMCs we confirmed that chloroquine both inhibited autophagy pathways and increased expression of BMPR-II protein via lysosomal inhibition. Consistent with the in vivo findings, chloroquine inhibited the proliferation and stimulated apoptosis of rat PASMCs in vitro, with no effect on endothelial cell proliferation or survival. Moreover, direct inhibition of autophagy pathways by ATG5 small interfering RNA knockdown inhibited proliferation of rat PASMCs. CONCLUSIONS Chloroquine and hydroxychloroquine exert beneficial effects in experimental PAH. The mechanism of action includes inhibition of autophagy pathways and inhibition of lysosomal degradation of BMPR-II.
Collapse
Affiliation(s)
- Lu Long
- Department of Medicine, Division of Respiratory Medicine, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
49
|
Seeger W, Pullamsetti SS. Mechanics and mechanisms of pulmonary hypertension-Conference summary and translational perspectives. Pulm Circ 2013; 3:128-36. [PMID: 23662189 PMCID: PMC3641720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany,Address correspondence to: Prof. Werner Seeger, Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Parkstrasse 1, 61231 Bad Nauheim, Germany
| | - Soni S. Pullamsetti
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
50
|
Seeger W, Pullamsetti SS. Mechanics and Mechanisms of Pulmonary Hypertension — Conference Summary and Translational Perspectives. Pulm Circ 2013. [DOI: 10.4103/2045-8932.109951] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The imagination has made more discoveries than the eye To catch an imagination of the future of pulmonary hypertension was exactly the spirit of the 55th ASPEN lung Conference. Basic scientists, pre-clinicians, clinicians and pharma joined together to achieve one goal—to combine creativity and inventiveness in a battle against a deadly disease. Summarizing this conference on “Mechanics and Mechanisms of Pulmonary Hypertension” is challenging in several aspects: To extract key novel findings from 12 state-of-the-art lectures, 25 oral presentations, 56 posters along with the integration of own data on discussed topics, to include hundreds of important questions, answers and discussion raised during the conference, to provide the line of thinking for the next 5–10 years of pulmonary hypertension (PH) research development and to focus equally well on both basic and translational research. Kurt Stenmark and Todd Bull, who chaired the conference, intensified this challenge several-fold by selecting a plethora of topics ranging from development of cardiopulmonary systems to pathogenesis of right ventricular failure, mechanics of right ventricle-pulmonary artery coupling to genomics and from understanding metabolic aspects to developing therapies for PH. With that, need not say, but a special admiration and thanks to the conference chairs for assembling such outstanding state-of-the-art speakers, for clustering the presentations logically and for leading lively and engaging discussions. Although it may look fragmentary, we would like to divide the conference summary into four major conceptual realms: The pulmonary vasculature in PH; right heart in PH; individualized approach- personalized medicine; and beyond PH-vascular abnormalities in COPD. - Joseph Joubert
Collapse
Affiliation(s)
- Werner Seeger
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Soni S. Pullamsetti
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Department of Lung Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|