1
|
Jin Y, Mao F, Wang X, Zhang J, Gao Y, Fan Y. The sonic hedgehog signaling inhibitor cyclopamine improves pulmonary arterial hypertension via regulating the bone morphogenetic protein receptor 2 pathway. Sci Rep 2025; 15:12512. [PMID: 40216933 PMCID: PMC11992095 DOI: 10.1038/s41598-025-97627-7] [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: 08/08/2024] [Accepted: 04/07/2025] [Indexed: 04/14/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease with hallmarks of pulmonary vascular remodeling and bone morphogenetic protein receptor 2 (BMPR2) mutation. Recent studies indicate Sonic hedgehog (SHH) signaling is involved in the proliferation of human pulmonary arterial smooth muscle cells (hPASMCs) but the role of the SHH signaling inhibitor cyclopamine in monocrotaline (MCT)-induced PAH has not been investigated. We hypothesized SHH promotes pulmonary vascular remodeling and that inhibition of SHH signaling by cyclopamine could attenuate pulmonary hypertension via the bone morphogenetic protein (BMP) pathway. SHH and BMPR2 proteins were measured in pulmonary arteries isolated from MCT-induced PAH rats and in hPASMCs. The therapeutic effects of cyclopamine were tested in PAH rats and in BMPR2 knockdown hPASMCs. SHH protein levels were increased in PAH rats and exogenous recombinant SHH protein promoted proliferation of hPASMCs via BMPR2 and osteopontin. Furthermore, cyclopamine attenuated hemodynamics and vascular remodeling via the BMP pathway in PAH rats. Finally, cyclopamine enhanced apoptosis and reduced proliferation in hPASMCs with impaired BMPR2. The findings of this study provide evidence that SHH has a role in pulmonary vascular remodeling via BMP4/BMPR2/ID1, and its inhibition by cyclopamine could be a potential therapeutic target in PAH.
Collapse
Affiliation(s)
- Youpeng Jin
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Fei Mao
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, PR China
| | - Xuehui Wang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Jie Zhang
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Yanting Gao
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China
| | - Youfei Fan
- Department of Pediatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwuweiqi Road, Jinan, 250014, Shandong, PR China.
| |
Collapse
|
2
|
Griffiths K, Grand RJ, Horan I, Certo M, Keeler RC, Mauro C, Tseng CC, Greig I, Morrell NW, Zanda M, Frenneaux MP, Madhani M. Fluorinated perhexiline derivative attenuates vascular proliferation in pulmonary arterial hypertension smooth muscle cells. Vascul Pharmacol 2024; 156:107399. [PMID: 38901807 DOI: 10.1016/j.vph.2024.107399] [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: 08/22/2023] [Revised: 04/30/2024] [Accepted: 05/26/2024] [Indexed: 06/22/2024]
Abstract
Increased proliferation and reduced apoptosis of pulmonary artery smooth muscle cells (PASMCs) is recognised as a universal hallmark of pulmonary arterial hypertension (PAH), in part related to the association with reduced pyruvate dehydrogenase (PDH) activity, resulting in decreased oxidative phosphorylation of glucose and increased aerobic glycolysis (Warburg effect). Perhexiline is a well-recognised carnitine palmitoyltransferase-1 (CPT1) inhibitor used in cardiac diseases, which reciprocally increases PDH activity, but is associated with variable pharmacokinetics related to polymorphic variation of the cytochrome P450-2D6 (CYP2D6) enzyme, resulting in the risk of neuro and hepatotoxicity in 'slow metabolisers' unless blood levels are monitored and dose adjusted. We have previously reported that a novel perhexiline fluorinated derivative (FPER-1) has the same therapeutic profile as perhexiline but is not metabolised by CYP2D6, resulting in more predictable pharmacokinetics than the parent drug. We sought to investigate the effects of perhexiline and FPER-1 on PDH flux in PASMCs from patients with PAH. We first confirmed that PAH PASMCs exhibited increased cell proliferation, enhanced phosphorylation of AKTSer473, ERK 1/2Thr202/Tyr204 and PDH-E1αSer293, indicating a Warburg effect when compared to healthy PASMCs. Pre-treatment with perhexiline or FPER-1 significantly attenuated PAH PASMC proliferation in a concentration-dependent manner and suppressed the activation of the AKTSer473 but had no effect on the ERK pathway. Perhexiline and FPER-1 markedly activated PDH (seen as dephosphorylation of PDH-E1αSer293), reduced glycolysis, and upregulated mitochondrial respiration in these PAH PASMCs as detected by Seahorse analysis. However, both perhexiline and FPER-1 did not induce apoptosis as measured by caspase 3/7 activity. We show for the first time that both perhexiline and FPER-1 may represent therapeutic agents for reducing cell proliferation in human PAH PASMCs, by reversing Warburg physiology.
Collapse
MESH Headings
- Cell Proliferation/drug effects
- Humans
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Perhexiline/pharmacology
- Perhexiline/analogs & derivatives
- Cells, Cultured
- Male
- Phosphorylation
- Female
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/pathology
- Middle Aged
- Signal Transduction/drug effects
- Antihypertensive Agents/pharmacology
- Adult
- Apoptosis/drug effects
- Case-Control Studies
Collapse
Affiliation(s)
- Kayleigh Griffiths
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Roger J Grand
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Ian Horan
- Department for Medicine, University of Cambridge, Cambridge, UK
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Ross C Keeler
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Claudio Mauro
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Chih-Chung Tseng
- Kosterlitz Centre for Therapeutics, University of Aberdeen, Aberdeen, UK
| | - Iain Greig
- Kosterlitz Centre for Therapeutics, University of Aberdeen, Aberdeen, UK
| | | | - Matteo Zanda
- The Institute of Chemical Sciences and Technologies, Milan, Italy
| | | | - Melanie Madhani
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
3
|
Yuan M, Liu T, Cai A, Zhan Z, Cheng Y, Wang Q, Xia Y, Shen N, Huang P, Zou X. Emerging connectivity of programmed cell death pathways and pulmonary vascular remodelling during pulmonary hypertension. J Cell Mol Med 2024; 28:e70003. [PMID: 39153207 PMCID: PMC11330287 DOI: 10.1111/jcmm.70003] [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: 10/25/2023] [Revised: 04/08/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024] Open
Abstract
Pulmonary hypertension (PH) is a chronic progressive vascular disease characterized by abnormal pulmonary vascular resistance and pulmonary artery pressure. The major structural alteration during PH is pulmonary vascular remodelling, which is mainly caused by the imbalance between proliferation and apoptosis of pulmonary vascular cells. Previously, it was thought that apoptosis was the only type of programmed cell death (PCD). Soon afterward, other types of PCD have been identified, including autophagy, pyroptosis, ferroptosis and necroptosis. In this review, we summarize the role of the above five forms of PCD in mediating pulmonary vascular remodelling, and discuss their guiding significance for PH treatment. The current review could provide a better understanding of the correlation between PCD and pulmonary vascular remodelling, contributing to identify new PCD-associated drug targets for PH.
Collapse
Affiliation(s)
- Meng‐nan Yuan
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Ting Liu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - An‐qi Cai
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Zibo Zhan
- School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yi‐li Cheng
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Qi‐yue Wang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Yu‐xuan Xia
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Nong‐er Shen
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| | - Xiao‐zhou Zou
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital, Affiliated People's HospitalHangzhou Medical CollegeHangzhouZhejiangChina
- Key Laboratory of Endocrine Gland Diseases of Zhejiang ProvinceZhejiang Provincial People's HospitalHangzhouZhejiangChina
| |
Collapse
|
4
|
Li W, Quigley K. Bone morphogenetic protein signalling in pulmonary arterial hypertension: revisiting the BMPRII connection. Biochem Soc Trans 2024; 52:1515-1528. [PMID: 38716930 PMCID: PMC11346422 DOI: 10.1042/bst20231547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/27/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and life-threatening vascular disorder, characterised by abnormal remodelling of the pulmonary vessels and elevated pulmonary artery pressure, leading to right ventricular hypertrophy and right-sided heart failure. The importance of bone morphogenetic protein (BMP) signalling in the pathogenesis of PAH is demonstrated by human genetic studies. Many PAH risk genes are involved in the BMP signalling pathway and are highly expressed or preferentially act on vascular endothelial cells. Endothelial dysfunction is recognised as an initial trigger for PAH, and endothelial BMP signalling plays a crucial role in the maintenance of endothelial integrity. BMPR2 is the most prevalent PAH gene, found in over 80% of heritable cases. As BMPRII protein is the major type II receptor for a large family of BMP ligands and expressed ubiquitously in many tissues, dysregulated BMP signalling in other cells may also contribute to PAH pathobiology. Sotatercept, which contains the extracellular domain of another transforming growth factor-β family type II receptor ActRIIA fused to immunoglobin Fc domain, was recently approved by the FDA as a treatment for PAH. Neither its target cells nor its mechanism of action is fully understood. This review will revisit BMPRII function and its extracellular regulation, summarise how dysregulated BMP signalling in endothelial cells and smooth muscle cells may contribute to PAH pathogenesis, and discuss how novel therapeutics targeting the extracellular regulation of BMP signalling, such as BMP9 and Sotatercept, can be related to restoring BMPRII function.
Collapse
Affiliation(s)
- Wei Li
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| | - Kate Quigley
- VPD Heart and Lung Research Institute, Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge CB2 0BB, U.K
| |
Collapse
|
5
|
Jiang Y, Song S, Liu J, Zhang L, Guo X, Lu J, Li L, Yang C, Fu Q, Zeng B. Epigenetic regulation of programmed cell death in hypoxia-induced pulmonary arterial hypertension. Front Immunol 2023; 14:1206452. [PMID: 37753070 PMCID: PMC10518698 DOI: 10.3389/fimmu.2023.1206452] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/28/2023] [Indexed: 09/28/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe progressive disease that may cause early right ventricular failure and eventual cardiac failure. The pathogenesis of PAH involves endothelial dysfunction, aberrant proliferation of pulmonary artery smooth muscle cells (PASMCs), and vascular fibrosis. Hypoxia has been shown to induce elevated secretion of vascular endothelial growth factor (VEGF), leading to the development of hypoxic PAH. However, the molecular mechanisms underlying hypoxic PAH remain incompletely understood. Programmed cell death (PCD) is a natural cell death and regulated by certain genes. Emerging evidence suggests that apoptotic resistance contributes to the development of PAH. Moreover, several novel types of PCD, such as autophagy, pyroptosis, and ferroptosis, have been reported to be involved in the development of PAH. Additionally, multiple diverse epigenetic mechanisms including RNA methylation, DNA methylation, histone modification, and the non-coding RNA molecule-mediated processes have been strongly linked to the development of PAH. These epigenetic modifications affect the expression of genes, which produce important changes in cellular biological processes, including PCD. Consequently, a better understanding of the PCD processes and epigenetic modification involved in PAH will provide novel, specific therapeutic strategies for diagnosis and treatment. In this review, we aim to discuss recent advances in epigenetic mechanisms and elucidate the role of epigenetic modifications in regulating PCD in hypoxia-induced PAH.
Collapse
Affiliation(s)
- Yuan Jiang
- College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Jingxin Liu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Liyuan Zhang
- Shanghai Baoxing Biological Equipment Engineering Co., Ltd, Shanghai, China
| | - Xiaofei Guo
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Jiayao Lu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Lie Li
- Shenzhen Reyson Biotechnology Co., Ltd, Shenzhen, China
- Nanjing Evertop Electronics Ltd., Nanjing, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Qiang Fu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Bin Zeng
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
6
|
Wang E, Zhou S, Zeng D, Wang R. Molecular regulation and therapeutic implications of cell death in pulmonary hypertension. Cell Death Discov 2023; 9:239. [PMID: 37438344 DOI: 10.1038/s41420-023-01535-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Pulmonary hypertension (PH) is a clinical and pathophysiological syndrome caused by changes in pulmonary vascular structure or function that results in increased pulmonary vascular resistance and pulmonary arterial pressure, and it is characterized by pulmonary endothelial dysfunction, pulmonary artery media thickening, pulmonary vascular remodeling, and right ventricular hypertrophy, all of which are driven by an imbalance between the growth and death of pulmonary vascular cells. Programmed cell death (PCD), different from cell necrosis, is an active cellular death mechanism that is activated in response to both internal and external factors and is precisely regulated by cells. More than a dozen PCD modes have been identified, among which apoptosis, autophagy, pyroptosis, ferroptosis, necroptosis, and cuproptosis have been proven to be involved in the pathophysiology of PH to varying degrees. This article provides a summary of the regulatory patterns of different PCD modes and their potential effects on PH. Additionally, it describes the current understanding of this complex and interconnected process and analyzes the therapeutic potential of targeting specific PCD modes as molecular targets.
Collapse
Affiliation(s)
- Enze Wang
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei, 230022, China
| | - Sijing Zhou
- Department of Occupational Disease, Hefei third clinical college of Anhui Medical University, Hefei, 230022, China
| | - Daxiong Zeng
- Department of pulmonary and critical care medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou, 215006, China.
| | - Ran Wang
- Department of respiratory and critical care medicine, the first affiliated hospital of Anhui medical university, Hefei, 230022, China.
| |
Collapse
|
7
|
Development and Optimisation of Inhalable EGCG Nano-Liposomes as a Potential Treatment for Pulmonary Arterial Hypertension by Implementation of the Design of Experiments Approach. Pharmaceutics 2023; 15:pharmaceutics15020539. [PMID: 36839861 PMCID: PMC9965461 DOI: 10.3390/pharmaceutics15020539] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/14/2022] [Accepted: 01/25/2023] [Indexed: 02/09/2023] Open
Abstract
Epigallocatechin gallate (EGCG), the main ingredient in green tea, holds promise as a potential treatment for pulmonary arterial hypertension (PAH). However, EGCG has many drawbacks, including stability issues, low bioavailability, and a short half-life. Therefore, the purpose of this research was to develop and optimize an inhalable EGCG nano-liposome formulation aiming to overcome EGCG's drawbacks by applying a design of experiments strategy. The aerodynamic behaviour of the optimum formulation was determined using the next-generation impactor (NGI), and its effects on the TGF-β pathway were determined using a cell-based reporter assay. The newly formulated inhalable EGCG liposome had an average liposome size of 105 nm, a polydispersity index (PDI) of 0.18, a zeta potential of -25.5 mV, an encapsulation efficiency of 90.5%, and a PDI after one month of 0.19. These results are in complete agreement with the predicted values of the model. Its aerodynamic properties were as follows: the mass median aerodynamic diameter (MMAD) was 4.41 µm, the fine particle fraction (FPF) was 53.46%, and the percentage of particles equal to or less than 3 µm was 34.3%. This demonstrates that the novel EGCG liposome has all the properties required to be inhalable, and it is expected to be deposited deeply in the lung. The TGFβ pathway is activated in PAH lungs, and the optimum EGCG nano-liposome inhibits TGFβ signalling in cell-based studies and thus holds promise as a potential treatment for PAH.
Collapse
|
8
|
CTRP9 Mitigates the Progression of Arteriovenous Shunt-Induced Pulmonary Artery Hypertension in Rats. Cardiovasc Ther 2021; 2021:4971300. [PMID: 34858521 PMCID: PMC8598355 DOI: 10.1155/2021/4971300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 12/30/2022] Open
Abstract
The present study is aimed at investigating the molecular mechanism of C1q/TNF-related protein 9 (CTRP9) and providing a new perspective in arteriovenous shunt-induced pulmonary arterial hypertension (PAH). PAH was established by an arteriovenous shunt placement performed in rats. Adenovirus(Ad)-CTRP9 and Ad-green fluorescent protein viral particles were injected into the rats through the tail vein. Following 12 weeks, the mean pulmonary arterial pressure (mPAP) and right ventricular systolic pressure (RVSP) were measured and morphological analysis was conducted to confirm the establishment of the PAH model. The systemic elevation of CTRP9 maintained pulmonary vascular homeostasis and protected the rats from dysfunctional and abnormal remodeling. CTRP9 attenuated the pulmonary vascular remodeling in the shunt group by decreasing the mPAP and RVSP, which was associated with suppressed inflammation, apoptosis, and extracellular matrix injury. In addition, CTRP9 dramatically increased the phosphorylation of AKT and p38-MAPK in the lung tissues of shunt-operated animals. These findings suggest a previously unrecognized effect of CTRP9 in pulmonary vascular homeostasis during PAH pathogenesis.
Collapse
|
9
|
Sitapara R, Lam TT, Gandjeva A, Tuder RM, Zisman LS. Phosphoproteomic analysis of lung tissue from patients with pulmonary arterial hypertension. Pulm Circ 2021; 11:20458940211031109. [PMID: 34966541 PMCID: PMC8711668 DOI: 10.1177/20458940211031109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 06/18/2021] [Indexed: 11/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disorder associated with high morbidity and mortality despite currently available treatments. We compared the phosphoproteome of lung tissue from subjects with idiopathic PAH (iPAH) obtained at the time of lung transplant with control lung tissue. The mass spectrometry-based analysis found 60,428 phosphopeptide features from which 6622 proteins were identified. Within the subset of identified proteins there were 1234 phosphopeptides with q < 0.05, many of which are involved in immune regulation, angiogenesis, and cell proliferation. Most notably there was a marked relative increase in phosphorylated (S378) IKZF3 (Aiolos), a zinc finger transcription factor that plays a key role in lymphocyte regulation. In vitro phosphorylation assays indicated that GSK3 alpha and/or GSK3 beta could phosphorylate IKZF3 at S378. Western blot analysis demonstrated increased pIKZF3 in iPAH lungs compared to controls. Immunohistochemistry demonstrated phosphorylated IKZF3 in lymphocytes surrounding severely hypertrophied pulmonary arterioles. In situ hybrization showed gene expression in lymphocyte aggregates in PAH samples. A BCL2 reporter assay showed that IKZF3 increased BCL2 promoter activity and demonstrated the potential role of phosphorylation of IKZF3 in the regulation of BCL mediated transcription. Kinase network analysis demonstrated potentially important regulatory roles of casein kinase 2, cyclin-dependent kinase 1 (CDK1), mitogen-associated protein kinases (MAPKs), and protein kinases (PRKs) in iPAH. Bioinformatic analysis demonstrated enrichment of RhoGTPase signaling and the potential importance of cGMP-dependent protein kinase 1 (PRKG). In conclusion, this unbiased phosphoproteomic analysis demonstrated several novel targets regulated by kinase networks in iPAH, and reinforced the potential role of immune regulation in the pathogenesis of iPAH. The identified up- and down-regulated phosphoproteins have potential to serve as biomarkers for PAH and to provide new insights for therapeutic strategies.
Collapse
Affiliation(s)
| | - TuKiet T Lam
- Department of Molecular Biophysics and Biochemistry, Yale University, Yale University, New Haven, CT, USA
- MS & Proteomics Resource, WM Keck Foundation Biotechnology Resource Laboratory, Yale University, New Haven, CT, USA
| | - Aneta Gandjeva
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Rubin M Tuder
- Program in Translational Lung Research, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lawrence S Zisman
- Rensselaer Center for Translational Research Inc., Troy, NY, USA
- Pulmokine Inc., Troy, NY, USA
| |
Collapse
|
10
|
Cai M, Li X, Dong H, Wang Y, Huang X. CCR7 and its related molecules may be potential biomarkers of pulmonary arterial hypertension. FEBS Open Bio 2021; 11:1565-1578. [PMID: 33630421 PMCID: PMC8167855 DOI: 10.1002/2211-5463.13130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/14/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a chronic progressive cardiovascular disease characterized by vascular remodeling and leading to right‐heart failure. The purpose of this research was to further study the pathogenesis of PAH and to detect potential prognostic signatures. Differentially expressed genes (DEGs) selected from GSE38267 were mostly enriched in inflammation‐related pathways, suggesting inflammation may be involved in the occurrence and development of PAH. Through the prediction and verification of related miRNAs and long noncoding RNAs using online databases and Gene Expression Omnibus (GEO) datasets, CCR7 and its related molecules, including hsa‐let‐7e‐5p and SNHG12, were identified as possible targets. The expression levels of CCR7, hsa‐let‐7e‐5p and SNHG12 were then verified by quantitative RT‐PCR in vivo and in vitro. Further study showed that silencing of SNHG12 decreased the expression of CCR7 under hypoxia treatment in vitro. Dual‐luciferase reporter assays were used to verify the relationship between hsa‐let‐7e‐5p and SNHG12. Collectively, our research reveals that a long noncoding RNA–miRNA–mRNA interaction network may be involved in the pathogenesis of PAH and suggests SNHG12, hsa‐let‐7e‐5p and CCR7 as potential biomarkers for PAH.
Collapse
Affiliation(s)
- Mengsi Cai
- Key Laboratory of Heart and Lung, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiuchun Li
- Key Laboratory of Heart and Lung, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Haoru Dong
- Key Laboratory of Heart and Lung, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Ying Wang
- Key Laboratory of Heart and Lung, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoying Huang
- Key Laboratory of Heart and Lung, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
11
|
Yang D, Yang X, Dai F, Wang Y, Yang Y, Hu M, Cheng Y. The Role of Bone Morphogenetic Protein 4 in Ovarian Function and Diseases. Reprod Sci 2021; 28:3316-3330. [PMID: 33966186 DOI: 10.1007/s43032-021-00600-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/22/2021] [Indexed: 12/19/2022]
Abstract
Bone morphogenetic proteins (BMPs) are the largest subfamily of the transforming growth factor-β (TGF-β) superfamily. BMP4 is a secreted protein that was originally identified due to its role in bone and cartilage development. Over the past decades, extensive literature has indicated that BMP4 and its receptors are widely expressed in the ovary. Dysregulation of BMP4 expression may play a vital role in follicular development, polycystic ovary syndrome (PCOS), and ovarian cancer. In this review, we summarized the expression pattern of BMP4 in the ovary, focused on the role of BMP4 in follicular development and steroidogenesis, and discussed the role of BMP4 in ovarian diseases such as polycystic ovary syndrome and ovarian cancer. Some studies have shown that the expression of BMP4 in the ovary is spatiotemporal and species specific, but the effects of BMP4 seem to be similar in follicular development of different species. In addition, BMP4 is involved in the development of hyperandrogenemia in PCOS and drug resistance in ovarian cancer, but further research is still needed to clarify the specific mechanisms.
Collapse
Affiliation(s)
- Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao Yang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Yang
- School of Physics & Technology, Key Laboratory of Artificial Micro/Nano Structure of Ministry of Education, Wuhan University, Wuhan, 430072, China.
| | - Min Hu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
12
|
Fazal S, Bisserier M, Hadri L. Molecular and Genetic Profiling for Precision Medicines in Pulmonary Arterial Hypertension. Cells 2021; 10:638. [PMID: 33805595 PMCID: PMC7999465 DOI: 10.3390/cells10030638] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare and chronic lung disease characterized by progressive occlusion of the small pulmonary arteries, which is associated with structural and functional alteration of the smooth muscle cells and endothelial cells within the pulmonary vasculature. Excessive vascular remodeling is, in part, responsible for high pulmonary vascular resistance and the mean pulmonary arterial pressure, increasing the transpulmonary gradient and the right ventricular "pressure overload", which may result in right ventricular (RV) dysfunction and failure. Current technological advances in multi-omics approaches, high-throughput sequencing, and computational methods have provided valuable tools in molecular profiling and led to the identification of numerous genetic variants in PAH patients. In this review, we summarized the pathogenesis, classification, and current treatments of the PAH disease. Additionally, we outlined the latest next-generation sequencing technologies and the consequences of common genetic variants underlying PAH susceptibility and disease progression. Finally, we discuss the importance of molecular genetic testing for precision medicine in PAH and the future of genomic medicines, including gene-editing technologies and gene therapies, as emerging alternative approaches to overcome genetic disorders in PAH.
Collapse
Affiliation(s)
| | | | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, New York, NY 10029, USA; (S.F.); (M.B.)
| |
Collapse
|
13
|
Su Z, Lin M, Zhang H, Li J, Wu M, Lv H, Wang J, Xie S. The Release of Cyclophilin A from Rapamycin-Stimulated Vascular Smooth Muscle Cells Mediated by Myosin II Activation: Involvement of Apoptosis but Not Autophagy. J Vasc Res 2020; 57:254-260. [PMID: 32526757 DOI: 10.1159/000506685] [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] [Received: 09/23/2019] [Accepted: 02/20/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The exocytosis of cyclophilin A (CyPA) by a vesicular pathway in response to reactive oxygen species has been determined. However, other sources of extracellular CyPA remain obscure. OBJECTIVE The aim of this study was to determine the role of autophagy in the secretion of CyPA. METHODS AND RESULTS Rapamycin induced the activation of autophagy and release of CyPA from primary cultured rat aortic smooth muscle cells (RASMCs). However, inhibition of autophagy by knockdown of Atg7 or chloroquine did not affect the rapamycin-induced release of CyPA. With the exception of myosin II activity, rho-associated coiled-coil kinase (ROCK), actin remodelling, and synaptic vesicles were not implicated in the release of rapamycin-induced CyPA. Finally, we confirmed that rapamycin-induced extracellular CyPA originated from apoptotic RASMCs. Furthermore, the decreased activation of myosin II by blebbistatin blocked the release of CyPA from apoptotic RASMCs induced by rapamycin. CONCLUSIONS Rapamycin induced the release of CyPA from apoptotic RASMCs but did not affect exocytosis through autophagosomes. ROCK, actin remodelling, and synaptic vesicles were not involved in the apoptosis-related release of CyPA. Myosin II activation modulated the apoptosis of vascular smooth muscle cells and the release of CyPA from rapamycin-induced apoptotic cell death.
Collapse
Affiliation(s)
- Zizhuo Su
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, and Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Maohuan Lin
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, and Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Haijun Zhang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jiajie Li
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, and Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Meiping Wu
- Department of Hematocyte Morphological Lab, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hanlu Lv
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, and Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Jingfeng Wang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, and Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China
| | - Shuanglun Xie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, and Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, China,
| |
Collapse
|
14
|
Morikawa M, Mitani Y, Holmborn K, Kato T, Koinuma D, Maruyama J, Vasilaki E, Sawada H, Kobayashi M, Ozawa T, Morishita Y, Bessho Y, Maeda S, Ledin J, Aburatani H, Kageyama R, Maruyama K, Heldin CH, Miyazono K. The ALK-1/SMAD/ATOH8 axis attenuates hypoxic responses and protects against the development of pulmonary arterial hypertension. Sci Signal 2019; 12:12/607/eaay4430. [PMID: 31719172 DOI: 10.1126/scisignal.aay4430] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dysregulated bone morphogenetic protein (BMP) signaling in endothelial cells (ECs) is implicated in vascular diseases such as pulmonary arterial hypertension (PAH). Here, we showed that the transcription factor ATOH8 was a direct target of SMAD1/5 and was induced in a manner dependent on BMP but independent of Notch, another critical signaling pathway in ECs. In zebrafish and mice, inactivation of Atoh8 did not cause an arteriovenous malformation-like phenotype, which may arise because of dysregulated Notch signaling. In contrast, Atoh8-deficient mice exhibited a phenotype mimicking PAH, which included increased pulmonary arterial pressure and right ventricular hypertrophy. Moreover, ATOH8 expression was decreased in PAH patient lungs. We showed that in cells, ATOH8 interacted with hypoxia-inducible factor 2α (HIF-2α) and decreased its abundance, leading to reduced induction of HIF-2α target genes in response to hypoxia. Together, these findings suggest that the BMP receptor type II/ALK-1/SMAD/ATOH8 axis may attenuate hypoxic responses in ECs in the pulmonary circulation and may help prevent the development of PAH.
Collapse
Affiliation(s)
- Masato Morikawa
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden.,Ludwig Institute for Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Yoshihide Mitani
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Katarina Holmborn
- Genome Engineering Zebrafish Facility, Science For Life Laboratory, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Taichi Kato
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Junko Maruyama
- Department of Anesthesiology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Eleftheria Vasilaki
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden.,Ludwig Institute for Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Hirofumi Sawada
- Department of Pediatrics, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan.,Department of Anesthesiology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Mai Kobayashi
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takayuki Ozawa
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasuyuki Morishita
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasumasa Bessho
- Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shingo Maeda
- Department of Medical Joint Materials, Kagoshima University, Kagoshima, Kagoshima 890-8544, Japan
| | - Johan Ledin
- Genome Engineering Zebrafish Facility, Science For Life Laboratory, Uppsala University, SE-752 36 Uppsala, Sweden
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology (RCAST), The University of Tokyo, Meguro-ku, Tokyo 153-8904, Japan
| | - Ryoichiro Kageyama
- Institute for Frontier Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazuo Maruyama
- Department of Anesthesiology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden. .,Ludwig Institute for Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan. .,Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-751 23 Uppsala, Sweden.,Ludwig Institute for Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-751 24 Uppsala, Sweden
| |
Collapse
|