1
|
Lei D, Liu Y, Liu Y, Jiang Y, Lei Y, Zhao F, Li W, Ouyang Z, Chen L, Tang S, Ouyang D, Li X, Li Y. The gut microbiota metabolite trimethylamine N-oxide promotes cardiac hypertrophy by activating the autophagic degradation of SERCA2a. Commun Biol 2025; 8:596. [PMID: 40210720 PMCID: PMC11986001 DOI: 10.1038/s42003-025-08016-9] [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: 09/07/2022] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
Trimethylamine oxide (TMAO) is a newly found intestinal microbiota metabolite. Here, we aimed to explore the effects of TMAO on calcium homeostasis and its implication in cardiac hypertrophy, especially focusing on the regulatory mechanism of TMAO on the key calcium transporter SERCA2a. Echocardiography and histological assessment showed that mice fed with TMAO or Choline for 8 weeks exhibited significant pathological changes of cardiac hypertrophy, which is accompanied by increased plasma levels of TMAO. The results indicated that TMAO could increase the intracellular Ca2+ level, up-regulate the expression of ANP and MYH7, and down-regulate SERCA2a expression, which could be reversed by overexpressing of SERCA2a and BAPTA-AM. Meanwhile, TMAO treatment promotes autophagy in vitro and in vivo. By employing immunofluorescence staining and immunoprecipitation assay, it was found that SERCA2a bound to ATG5 and transported to autophagosomes via the ATG5 complex for degradation under TMAO conditions. Furthermore, either 3MA or siATG5 could ameliorate TMAO-induced cardiomyocyte hypertrophy and SERCA2a degradation. Finally, in vivo intervention showed that 3MA could relieve cardiac hypertrophy and rescue the down-regulation of SERCA2a in TMAO-fed mice. The current study identifies a mechanism in which TMAO promotes cardiac hypertrophy via elevated intracellular Ca2+ levels and enhanced autophagy degradation of SERCA2a.
Collapse
Affiliation(s)
- Dongyu Lei
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Department of Physiology, School of Basic Medicine, Xinjiang Medical University, Urumqi, 830017, China
| | - Yi Liu
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yuan Liu
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, China
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Yujie Jiang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yuyan Lei
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Phase I Clinical Trial Laboratory, the Second Nanning People's Hospital, Guangxi, China
| | - Feilong Zhao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Zhonghua Ouyang
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, 411000, China
| | - Lulu Chen
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, 411000, China
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha, 410000, China
| | - Dongsheng Ouyang
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, 411000, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, 411000, China.
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| |
Collapse
|
2
|
Cabiati M, Biondi F, Ghelardoni S, Casieri V, Lionetti V, Sgalippa A, Del Ry S, Madonna R. Ambrisentan Retains Its Pro-Autophagic Activity on Human Pulmonary Artery Endothelial Cells Exposed to Hypoxia in an In Vitro Model Mimicking Diabetes. J Cell Mol Med 2025; 29:e70528. [PMID: 40205749 PMCID: PMC11982177 DOI: 10.1111/jcmm.70528] [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: 01/08/2025] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/11/2025] Open
Abstract
Cardiovascular comorbidities are associated with reduced treatment response in group 1 pulmonary arterial hypertension (PAH). This may result from misdiagnosis of group 2 PH, but it can also be explained as the loss of ability of pulmonary endothelial cells to respond to specific antiremodeling drugs. We evaluated the effects of high glucose (HG) and hyperosmolar stress (high mannitol, HM) on the response of human pulmonary artery endothelial cells (hPAECs) to ambrisentan (AMB), focusing on autophagy, viability, apoptosis and several microRNAs involved in pulmonary arterial remodelling. hPAECs were incubated with 30.5 mM HG or 25 mM HM, with/without 0.02 nM AMB in normoxia (Nx) or hypoxia (Hx) for 24 h. Hx reduced cell survival (p = 0.03) and autophagy (p = 0.02), an effect mimicked by HG and HM only in Nx. In Nx and Hx, AMB reverted the effect of HG, but not HM on autophagy, almost completely or partially, respectively. Compared to Nx, Hx increased the antiapoptotic miR124-3p in vehicle-treated hPAEC (p = 0.002), and induced an opposite effect on antiapoptotic and proliferative miR191-3p. In Nx, AMB induced miR124-3p in HG- (p = 0.04 vs. HG+A_Nx) and HM-treated (p < 0.0001 vs. HM+AMB_Nx) hPAECs, and miR191-3p in HM-treated hPAECs (p = 0.03). In H, A induced a similar effect on miR124-3p in hPAEC exposed to AMB+HM (p = 0.02). In hPAEC exposed to Hx, AMB retains its pro-autophagic effects in an in vitro model mimicking diabetes. miR124-3p and, to a lesser extent, miR191-3p may act as biomarkers of disease and treatment response to specific drugs in patients with PAH and diabetes.
Collapse
Affiliation(s)
- Manuela Cabiati
- Laboratory of Biochemistry and Molecular BiologyInstitute of Clinical Physiology, CNRPisaItaly
| | - Filippo Biondi
- Department of Pathology, Cardiology DivisionUniversity of PisaPisaItaly
| | - Sandra Ghelardoni
- Department of Pathology, Laboratory of BiochemistryUniversity of PisaPisaItaly
| | - Valentina Casieri
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical SciencesInterdisciplinary Research Center “Health Science,” Scuola Superiore Sant'AnnaPisaItaly
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical SciencesInterdisciplinary Research Center “Health Science,” Scuola Superiore Sant'AnnaPisaItaly
| | - Agnese Sgalippa
- Laboratory of Biochemistry and Molecular BiologyInstitute of Clinical Physiology, CNRPisaItaly
| | - Silvia Del Ry
- Laboratory of Biochemistry and Molecular BiologyInstitute of Clinical Physiology, CNRPisaItaly
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical SciencesInterdisciplinary Research Center “Health Science,” Scuola Superiore Sant'AnnaPisaItaly
| | - Rosalinda Madonna
- Laboratory of Biochemistry and Molecular BiologyInstitute of Clinical Physiology, CNRPisaItaly
- Department of Pathology, Cardiology DivisionUniversity of PisaPisaItaly
| |
Collapse
|
3
|
Milano G, Reinero M, Puyal J, Tozzi P, Samaja M, Porte-Thomé F, Beghetti M. Inhibition of Sodium/Hydrogen Exchanger-1 in the Right Ventricle and Lung Dysfunction Induced by Experimental Pulmonary Arterial Hypertension in Rats. J Am Heart Assoc 2025; 14:e036859. [PMID: 40055146 DOI: 10.1161/jaha.124.036859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/06/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Life-threatening pulmonary arterial hypertension (PAH) still lacks a direct therapeutic approach targeted to the molecular defects associated with the disease. Here, we focus on the impaired regulation of intracellular acidity and sodium/calcium overload by testing the hypothesis that inhibiting NHE-1 (sodium/hydrogen exchanger isoform 1) with rimeporide enables the recovery of pulmonary and right ventricular dysfunctions in the Sugen5416/hypoxia PAH model in rats. METHODS AND RESULTS Adult Sprague-Dawley male rats (n=44) rats were divided into 2 broad groups: control and Sugen5416/hypoxia. After verifying PAH insurgence in the Sugen5416/hypoxia group by transthoracic echocardiography and pulse-wave Doppler analysis, rats were treated with either 100 mg/kg per day rimeporide or placebo in drinking water for 3 weeks. The functional, morphological (fibrosis and hypertrophy), and biochemical (inflammation, signaling pathways) dysfunctions caused by PAH were partially reverted by rimeporide in both the lungs and myocardium, where the most striking effects were observed in the right ventricle. Rimeporide improved hemodynamics in the pulmonary circulation and in the right ventricle, with decrease in right ventricle hypertrophy, pulmonary vascular remodeling, inflammation, and fibrosis. No effect of rimeporide was detected in control rats. The protective effect of rimeporide was accompanied by decreased p-Akt/Akt (phosphorylated protein kinase B/protein kinase B) ratio and increased autophagy flux mainly in the right ventricle. CONCLUSIONS By specifically inhibiting NHE-1, rimeporide at the selected dosage revealed remarkable anti-PAH effects by preventing the functional, morphological, and biochemical deleterious effects of PAH on the right ventricle and lungs. Rimeporide should be considered as a potential treatment for PAH.
Collapse
MESH Headings
- Animals
- Rats, Sprague-Dawley
- Male
- Disease Models, Animal
- Ventricular Dysfunction, Right/metabolism
- Ventricular Dysfunction, Right/physiopathology
- Ventricular Dysfunction, Right/etiology
- Ventricular Dysfunction, Right/drug therapy
- Ventricular Dysfunction, Right/prevention & control
- Sodium-Hydrogen Exchanger 1/antagonists & inhibitors
- Sodium-Hydrogen Exchanger 1/metabolism
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/complications
- Ventricular Function, Right/drug effects
- Rats
- Lung/drug effects
- Lung/metabolism
- Lung/physiopathology
- Pyrroles
- Indoles
- Heart Ventricles/metabolism
- Heart Ventricles/drug effects
- Heart Ventricles/physiopathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/physiopathology
- Guanidines/pharmacology
- Hypertrophy, Right Ventricular/physiopathology
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/prevention & control
- Hypertrophy, Right Ventricular/etiology
Collapse
Affiliation(s)
- Giuseppina Milano
- Department Cœur-Vaisseaux, Cardiac Surgery Center University Hospital of Lausanne Switzerland
| | - Melanie Reinero
- Department Cœur-Vaisseaux, Cardiac Surgery Center University Hospital of Lausanne Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences University of Lausanne Switzerland
- CURML, University Center of Legal Medicine, Lausanne University Hospital Lausanne Switzerland
| | - Piergiorgio Tozzi
- Department Cœur-Vaisseaux, Cardiac Surgery Center University Hospital of Lausanne Switzerland
| | | | | | - Maurice Beghetti
- Unité de Cardiologie Pédiatrique University Hospital of Geneva, University of Geneva Switzerland
| |
Collapse
|
4
|
Tan W, Wang Y, Li M, Zhao C, Hu Y, Gao R, Chen Z, Hu L, Li Q. A novel pyridine-2-one AMPK inhibitor: Discovery, mechanism, and in vivo evaluation in a hypoxic pulmonary arterial hypertension rat model. Eur J Med Chem 2025; 286:117266. [PMID: 39826489 DOI: 10.1016/j.ejmech.2025.117266] [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: 11/02/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
AMP-activated protein kinase (AMPK), a heterotrimeric serine-threonine kinase, has been identified as a promising target for regulating vascular remodeling in pulmonary arterial hypertension (PAH) due to its capacity to promote proliferation, autophagy, and anti-apoptosis in pulmonary artery smooth muscle cells (PASMCs). However, research into AMPK inhibitors is very limited. Herein, a virtual screening strategy was employed to identify CHEMBL3780091 as a lead compound for a series of novel AMPK inhibitors by exploring the structure-activity relationship around a specific pyridine-2-one scaffold. Subsequently, the most promising 13a was observed to exhibit excellent AMPK inhibitory activity and favorable anti-proliferative activity against PASMCs through the inhibition of the AMPK signaling pathway in vitro. Moreover, compound 13a significantly reduced right ventricular systolic pressure, attenuated vascular remodeling, and improved right heart function in hypoxia-induced PAH rats in vivo. In conclusion, this study provides a novel and potential lead compound for the study of AMPK inhibitors and a new direction for the development of PAH drugs that focus on improving vascular remodeling.
Collapse
Affiliation(s)
- Wenhua Tan
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yu Wang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Mengqi Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Congke Zhao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yuanbo Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Ruizhe Gao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China.
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
5
|
Wang Y, Wang Y, Zhang W. Dysregulation of Mitochondrial in Pulmonary Hypertension-Related Right Ventricular Remodeling: Pathophysiological Features and Targeting Drugs. Rev Cardiovasc Med 2025; 26:25781. [PMID: 40160582 PMCID: PMC11951289 DOI: 10.31083/rcm25781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 04/02/2025] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening condition characterized by right ventricular (RV) remodeling, which is a major determinant of patient survival. The progression of right ventricular remodeling is significantly influenced by mitochondrial dysfunction, providing profound insights into vascular health and cardiovascular risk. In this review, we discuss the molecular targets, pathophysiological characteristics, and potential mechanisms underlying mitochondrial dysfunction in PH, encompassing disturbances in mitochondrial dynamics, inflammation, and dysregulation of mitochondrial energy metabolism. Finally, we review the primary therapeutic targets currently utilized to address cardiac dysfunction resulting from mitochondrial damage. Hopefully, this might inspire novel approaches to the management of cardiovascular disorders.
Collapse
Affiliation(s)
- Yuehan Wang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Yingzhuo Wang
- The First Clinical Medical College, Nanchang University, 330036 Nanchang, Jiangxi, China
| | - Weifang Zhang
- Departments of Pharmacy, The Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China
- Huankui Academy, Nanchang University, 330036 Nanchang, Jiangxi, China
| |
Collapse
|
6
|
El-Ashmawy NE, Al-Ashmawy GM, Kamel AA, Khedr EG. Unlocking the therapeutic potential of canagliflozin in NAFLD: Insights into AMPK/SIRT1-mediated lipophagy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167666. [PMID: 39837063 DOI: 10.1016/j.bbadis.2025.167666] [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: 11/13/2024] [Revised: 12/23/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a rising global health problem. The antidiabetic canagliflozin (CANA) has been proposed to ameliorate the metabolic abnormalities in NAFLD. AIM This study aimed to explore the possible anti-NAFLD effects of CANA in rats and HepG2 cells, focusing on AMPK/SIRT1-mediated lipophagy. METHODS Wistar rats were assigned to four groups: control group, NAFLD group, NAFLD+CANA group, and NAFLD+CANA+chloroquine (CQ) group, where CQ served as autophagy inhibitor. HepG2 cells were also divided into four groups: control group, NAFLD group, NAFLD+CANA group, and NAFLD+CANA+compound C (Comp C) group, where Comp C served as AMPK inhibitor. RESULTS The histopathological examination showed that CANA alleviated hepatic and intracellular lipid deposition in rats and HepG2 cells. CANA induced lipophagy by increasing LC3-II levels and lowering both p62 and perilipin 2 levels in rats and HepG2 cells, in addition to decreasing mTOR protein expression in rats' livers. These outcomes were associated with upregulation of the lipophagy regulator Rab7 and downregulation of the ER stress-related protein CHOP. CANA enhanced autophagic engulfment of lipid droplets while decreased ER stress and mitochondrial damage in rats' livers, as demonstrated by TEM. In rats, CANA improved hyperglycemia, hyperinsulinemia, dyslipidemia, and obesity. In HepG2 cells, CANA's effects were linked to increased phosphorylated AMPK level and enhanced SIRT1 level and expression. However, blocking lipophagy in rats and AMPK in HepG2 cells markedly weakened CANA's protective effects against NAFLD. CONCLUSION CANA ameliorated NAFLD via enhancing AMPK/SIRT1-mediated lipophagy, suggesting its potential as a therapeutic intervention for this metabolic disorder.
Collapse
Affiliation(s)
- Nahla E El-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt; Department of Pharmacology & Biochemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk, Egypt
| | - Ghada M Al-Ashmawy
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt; Department of Biochemistry, Faculty of Pharmacy, Alsalam University in Egypt, Egypt
| | - Asmaa A Kamel
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt.
| | - Eman G Khedr
- Department of Biochemistry, Faculty of Pharmacy, Tanta University, Egypt
| |
Collapse
|
7
|
Guglielmi G, Dimopoulos K, Wort SJ. New therapies in pulmonary arterial hypertension: Recent insights. INTERNATIONAL JOURNAL OF CARDIOLOGY CONGENITAL HEART DISEASE 2025; 19:100571. [PMID: 39991439 PMCID: PMC11847046 DOI: 10.1016/j.ijcchd.2025.100571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Pulmonary Arterial Hypertension (PAH) is a complex and progressive disease characterized by elevated pulmonary vascular resistance and right heart failure. Current therapies primarily focus on pulmonary vasodilation; however, novel approaches that target the underlying pathophysiological mechanisms-such as TGF-β signalling, epigenetic alterations, growth factors, inflammation, and extracellular matrix remodelling-are promising alternatives for improving treatment outcomes. This is a review of recent advances in the development of innovative therapeutic strategies for PAH. The first section of this paper explores approaches targeting TGF-β signalling, both acting directly on receptors through drugs like Sotatercept and exogenous BMP9, and indirectly, inhibiting the degradation of key receptors, such as BMPR2. Subsequent sections describe treatments that target epigenetic regulators, e.g. poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors and direct BRD4 antagonists, tyrosine kinase inhibitors (Seralutinib), and therapies aimed at inflammation, such as IL-6 inhibitors, CD-20 inhibitors, and monoclonal antibodies that prevent macrophage migration. Finally, strategies that target the serotonin pathway, and other metabolic and hormonal pathways are described. This review includes both preclinical and clinical trial data that support efficacy, safety and the future potential of such therapies. Collectively, these therapeutic approaches can be valuable in treating PAH by targeting multiple aspects of its pathogenesis, potentially resulting in improved clinical outcomes for patients affected by this debilitating, life-limiting condition.
Collapse
Affiliation(s)
- Giulia Guglielmi
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, United Kingdom
| | - Konstantinos Dimopoulos
- Adult Congenital Heart Centre and National Centre for Pulmonary Hypertension, Royal Brompton Hospital, London, United Kingdom
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - S. John Wort
- National Pulmonary Hypertension Service, Royal Brompton Hospital, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Liang Y, Ornatowski W, Lu Q, Sun X, Yegambaram M, Feng A, Dong Y, Aggarwal S, Unwalla HJ, Fineman JR, Black SM, Wang T. Chloroquine Restores eNOS Signaling in Shunt Endothelial Cells via Inhibiting eNOS Uncoupling. Int J Mol Sci 2025; 26:1352. [PMID: 39941119 PMCID: PMC11818845 DOI: 10.3390/ijms26031352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by increased lung vascular stiffness and impaired vessel relaxation, primarily due to reduced nitric oxide (NO) production in endothelial cells. Recent studies indicate that chloroquine, an autophagy inhibitor, may help lower pulmonary arterial pressure and enhance lung vascular function. This study investigates the mechanisms underlying the chloroquine-mediated restoration of NO bioavailability in endothelial cells derived from aortopulmonary shunt lambs, a relevant model for congenital heart defect (CHD)-associated PAH. We found that NO production was significantly reduced in shunt pulmonary artery endothelial cells (PAECs), attributable to decreased levels of tetrahydrobiopterin (BH4) and diminished expression of GTP cyclohydrolase 1 (GCH1), despite a slight increase in endothelial nitric oxide synthase (eNOS) levels. Chloroquine robustly restored endothelial NO production, which correlated with increased BH4 levels and restored GCH1 expression. The mechanistically upregulated carboxyl terminus of Hsp70-interacting protein (CHIP) in shunt PAECs is responsible for heightened GCH1 degradation, and chloroquine disrupted the assembly of the GCH1-HSP70-CHIP complex to preserve cellular GCH1. Similarly, another autophagy inhibitor, bafilomycin A1, demonstrated comparable effects. These findings suggest that autophagy inhibition can effectively enhance NO synthesis in endothelial cells experiencing depleted NO bioavailability, presenting a potential therapeutic strategy for managing PAH.
Collapse
Affiliation(s)
- Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Wojciech Ornatowski
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Hoshang J. Unwalla
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
9
|
Wang X, Huang X, Zhang Y, Huo H, Zhou G, Shen L, Li L, He B. Hydrogen sulfide attenuates disturbed flow-induced vascular remodeling by inhibiting LDHB-mediated autophagic flux. Redox Biol 2025; 79:103456. [PMID: 39647238 PMCID: PMC11666931 DOI: 10.1016/j.redox.2024.103456] [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: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024] Open
Abstract
Disturbed flow (DF) plays a critical role in the development and progression of cardiovascular disease (CVD). Hydrogen sulfide (H2S) is involved in physiological processes within the cardiovascular system. However, its specific contribution to DF-induced vascular remodeling remains unclear. Here, we showed that the H2S donor, NaHS suppressed DF-induced vascular remodeling in mice. Further experiments demonstrated that NaHS inhibited the proliferation and migration of vascular smooth muscle cells (VSMCs) induced by platelet-derived growth factor-BB (PDGF), as well as the autophagy triggered by DF and PDGF. Mechanistically, RNA-Seq results revealed that NaHS counteracted the PDGF-induced upregulation of lactate dehydrogenase B (LDHB). Overexpression of LDHB abolished the protective effect of NaHS on DF-induced vascular remodeling. Furthermore, LDHB interacted with vacuolar-type proton ATPase catalytic subunit A (ATP6V1A), leading to lysosomal acidification, a process that was attenuated by NaHS treatment. The residues of leucine (Leu) 57 in ATP6V1A and serine (Ser) 269 in LDHB are critical for their interaction. Notably, the expression of LDHB was found to be elevated in vascular tissues from patients with abdominal aortic aneurysms (AAA) and thoracic aortic aneurysms (TAA). These data identify a molecular mechanism by which H2S attenuates DF-induced vascular remodeling by inhibiting LDHB and disrupting the interaction between LDHB and ATP6V1A, thereby impeding the autophagy process. Our findings provide insight that H2S or targeting LDHB has therapeutic potential for preventing and treating vascular remodeling.
Collapse
Affiliation(s)
- Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xiying Huang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yongya Zhang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Huanhuan Huo
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Guo Zhou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Long Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, China.
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China.
| |
Collapse
|
10
|
Alves-Silva JM, Zuzarte M, Marques C, Rodrigues T, Barbeitos J, Caetano R, Baptista R, Salgueiro L, Girão H. 1,8-Cineole reduces pulmonary vascular remodelling in pulmonary arterial hypertension by restoring intercellular communication and inhibiting angiogenesis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 137:156334. [PMID: 39813848 DOI: 10.1016/j.phymed.2024.156334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 12/02/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
BACKGROUND Pulmonary Arterial Hypertension (PAH) is characterized by pulmonary vascular remodelling, often associated with disruption of BMPR2/Smad1/5 and BMPR2/PPAR-γ signalling pathways that ultimately lead to right ventricle failure. Disruption of intercellular junctions and communication and a pro-angiogenic environment are also characteristic features of PAH. Although, current therapies improve pulmonary vascular tone, they fail to tackle other key pathological features that could prevent disease progression. In this scenario, aromatic plants emerge as promising sources of bioactive compounds, with 1,8-cineole standing out due to its hypotensive properties and cardioprotective effect in PAH. PURPOSE The present study aims to explore for the first time the effect of 1,8-cineole in pulmonary vascular remodelling associated with PAH. METHODS Resorting to the monocrotaline (MCT)-induced PAH animal model, the effect of 1,8-cineole on vascular remodelling including interstitial collagen accumulation, smooth muscle cell proliferation and protein levels of BMPR2 pathway-related proteins, was assessed by microscopy and western blot (WB) analysis. The integrity of gap junctions, pulmonary surfactant, mitochondrial structure and endothelial cell barrier were evaluated by transmission electron microscopy, confocal microscopy and WB analysis. Furthermore, the effect of 1,8-cineole on angiogenesis was determined on pulmonary artery endothelial cells (PAEC) submitted to hypoxia using the scratch wound and Matrigel angiogenesis assays, and the number of sprouts on isolated healthy and diseased pulmonary artery rings, treated with the compound, enabled the validation of these effects. RESULTS 1,8-Cineole mitigated PAH-associated derailment of both BMPR2/Smad1/5 and BMPR2/PPAR-γ pathways and concomitantly reduced interstitial fibrosis and the arterial medial layer thickness in pulmonary arteries. The compound restored gap junction, lung surfactant and mitochondrial integrity and preserved endothelial barrier integrity. Furthermore, 1,8-cineole exerted an anti-angiogenic effect, by impairing the formation of vessel-like structures in PAEC and sprouting formation in isolated pulmonary arteries. CONCLUSION The present study brings new insights about the mechanisms whereby 1,8-cineole impacts pulmonary vascular remodelling and demonstrates the potential of 1,8-cineole as a therapeutic strategy to hamper PAH progression.
Collapse
Affiliation(s)
- Jorge M Alves-Silva
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Mónica Zuzarte
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal; Univ Coimbra, Chemical Engineering and Renewable Resources for Sustainability (CERES), Department of Chemical Engineering, Coimbra 3030-790, Portugal.
| | - Carla Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Teresa Rodrigues
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Júlia Barbeitos
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, Coimbra 3000-548, Portugal
| | - Rui Caetano
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Centro de Anatomia Patológica Germano de Sousa, Coimbra 3000-377, Portugal; Centre of Investigation on Genetics and Oncobiology (CIMAGO), Azinhaga de S. Comba, 3000-548 Coimbra, Portugal
| | - Rui Baptista
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal; Cardiology Department, Centro Hospitalar de Entre o Douro e Vouga, Santa Maria da Feira 4520-211, Portugal
| | - Lígia Salgueiro
- Univ Coimbra, Faculty of Pharmacy, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Chemical Engineering and Renewable Resources for Sustainability (CERES), Department of Chemical Engineering, Coimbra 3030-790, Portugal
| | - Henrique Girão
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Azinhaga de S. Comba, Coimbra 3000-548, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| |
Collapse
|
11
|
Harvey LD, Alotaibi M, Tai YY, Tang Y, Kim HJJ, Kelly NJ, Sun W, Woodcock CSC, Arshad S, Culley MK, El Khoury W, Xie R, Al Aaraj Y, Zhao J, Hafeez N, Rao RJ, Jiang S, Negi V, Kirillova A, Perk D, Watson AM, St Croix CM, Stolz DB, Lee JY, Cheng MH, Zhang M, Detmer S, Guzman E, Manan RS, Saggar R, Haley KJ, Waxman AB, Okawa S, Schwantes-An TH, Pauciulo MW, Wang B, Webb A, Chauvet C, Anderson DG, Nichols WC, Desai AA, Lafyatis R, Nouraie SM, Wu H, McDonald JG, Cheng S, Bahar I, Bertero T, Benza RL, Jain M, Chan SY. Lysosomal dysfunction and inflammatory sterol metabolism in pulmonary arterial hypertension. Science 2025; 387:eadn7277. [PMID: 39847635 DOI: 10.1126/science.adn7277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025]
Abstract
Vascular inflammation regulates endothelial pathophenotypes, particularly in pulmonary arterial hypertension (PAH). Dysregulated lysosomal activity and cholesterol metabolism activate pathogenic inflammation, but their relevance to PAH is unclear. Nuclear receptor coactivator 7 (NCOA7) deficiency in endothelium produced an oxysterol and bile acid signature through lysosomal dysregulation, promoting endothelial pathophenotypes. This oxysterol signature overlapped with a plasma metabolite signature associated with human PAH mortality. Mice deficient for endothelial Ncoa7 or exposed to an inflammatory bile acid developed worsened PAH. Genetic predisposition to NCOA7 deficiency was driven by single-nucleotide polymorphism rs11154337, which alters endothelial immunoactivation and is associated with human PAH mortality. An NCOA7-activating agent reversed endothelial immunoactivation and rodent PAH. Thus, we established a genetic and metabolic paradigm that links lysosomal biology and oxysterol processes to endothelial inflammation and PAH.
Collapse
Affiliation(s)
- Lloyd D Harvey
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mona Alotaibi
- Division of Pulmonary and Critical Care Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Yi-Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hee-Jung J Kim
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neil J Kelly
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chen-Shan C Woodcock
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sanya Arshad
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Miranda K Culley
- Medical Scientist Training Program, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rong Xie
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Neha Hafeez
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rashmi J Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Siyi Jiang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anna Kirillova
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dror Perk
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Annie M Watson
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Donna B Stolz
- Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ji Young Lee
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Mary Hongying Cheng
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Manling Zhang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Cardiology, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Samuel Detmer
- Department of Chemistry, Massachusetts Institute of Technology, Boston, MA, USA
| | - Edward Guzman
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Rajith S Manan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Rajan Saggar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
- Department of Pathology, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Kathleen J Haley
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Okawa
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tae-Hwi Schwantes-An
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Michael W Pauciulo
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Bing Wang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amy Webb
- Department of Biomedical Informatics, Ohio State University, Columbus, OH, USA
| | - Caroline Chauvet
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Sophia-Antipolis, Valbonne, France
| | - Daniel G Anderson
- Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Boston, MA, USA
| | - William C Nichols
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ankit A Desai
- Department of Medicine, Indiana University, Indianapolis, IN, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - S Mehdi Nouraie
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Haodi Wu
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jeffrey G McDonald
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ivet Bahar
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), Sophia-Antipolis, Valbonne, France
| | - Raymond L Benza
- Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohit Jain
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh, Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Cardiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Li H, Li X, Sun Y, Zhi Z, Song L, Li M, Feng Y, Zhang Z, Liu Y, Chen Y, Zhao F, Zhu T. The Role of Ion Channels in Pulmonary Hypertension: A Review. Pulm Circ 2025; 15:e70050. [PMID: 39958971 PMCID: PMC11830494 DOI: 10.1002/pul2.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/16/2024] [Accepted: 01/31/2025] [Indexed: 02/18/2025] Open
Abstract
Pulmonary hypertension (PH) constitutes a critical challenge in cardiopulmonary medicine with a pathogenesis that is multifaceted and intricate. Ion channels, crucial determinants of cellular electrochemical gradient modulation, have emerged as significant participants in the pathophysiological progression of PH. These channels, abundant on the membranes of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs), pivotally navigate the nuanced interplay of cell proliferation, migration, and endothelial function, each vital to the pulmonary vascular remodeling (PVR) hallmark of PH. Our review delves into the mechanistic insights of potassium, calcium, magnesium, zinc, and chloride ion channels in relation to their involvement in PH. It not only emphasizes the notable advances and discoveries that cast these ion channels as underlying factors in the etiology and exacerbation of PH but also highlights their potential as innovative therapeutic targets.
Collapse
Affiliation(s)
- Han‐Fei Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Xin‐Yao Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yu‐Qing Sun
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Ze‐Ying Zhi
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Liao‐Fan Song
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Meng Li
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yi‐Ming Feng
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Zhi‐Hao Zhang
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yan‐Feng Liu
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Yu‐Jing Chen
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Fan‐Rong Zhao
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
| | - Tian‐Tian Zhu
- College of PharmacyXinxiang Medical UniversityXinxiangChina
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug InterventionXinxiangChina
- Department of PharmacyThe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| |
Collapse
|
13
|
Li C, Li C, Jiang Y, Liu M, Yang C, Lu J, Jiang Y. Hypoxia-induced TPC2 transcription and glycosylation aggravates pulmonary arterial hypertension by blocking autophagy flux. Sci Rep 2024; 14:31223. [PMID: 39732974 DOI: 10.1038/s41598-024-82552-y] [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: 02/05/2024] [Accepted: 12/06/2024] [Indexed: 12/30/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a serious medical condition that causes a failure in the right heart. Two-pore channel 2 (TPC2) is upregulated in PAH, but its roles in PAH remain largely unknown. Our investigation aims at the mechanisms by which TPC2 regulates PAH development. We established an experimental PAH rat model via monocrotaline administration. Human and rat pulmonary arterial smooth muscle cells (PASMCs) were treated hypoxia as in vitro cell PAH models. The thickness of pulmonary arterial wall and obstructive arteriopathy in rats were examined. Autophagy was detected through TEM, and Ca2+ measurement and mRFP-GFP-LC3 transfection. The expression of α-SMA, LC3, p62, TPC2, HIF1α and STT3B were analyzed by qRT-PCR, western blot or IHC staining. The binding of HIF1α to TPC2 promoter was determined by ChIP-qPCR and EMSA assays. TPC2 glycosylation was evaluated by western blot. Transwell assay was applied to analyze cell migration. TPC2 expression was promoted and autophagy was inhibited in PAH rats and hypoxia-treated PASMCs. HIF1α directly bound to the promoter of TPC2, thus transcriptionally activating its expression in PAH rats and hypoxic PASMCs. Knockdown of TPC2 facilitated autophagic flux and repressed PASMC migration. STT3B enhanced TPC2 glycosylation in hypoxic PASMCs. Furthermore, Overexpression of TPC2 suppressed autophagic flux and promoted PASMC migration, but these effects were abrogated by STT3B knockdown or PNGase F, an eraser of N-linked glycans. Suppression of TPC2 enhanced autophagy and alleviated PAH in vivo. HIF1α-induced TPC2 transcription and subsequent STT3B-dependent TPC2 glycosylation inhibit autophagic flux and aggravate PAH. Our study suggests TCP2 as a potential therapeutic target for PAH.
Collapse
Affiliation(s)
- Chao Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China
| | - Cheng Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China
| | - YuFei Jiang
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China
- Faculty of Healthy Science, University of Macau, Macau, 999078, China
| | - MoFei Liu
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China
| | - ChengYi Yang
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China
| | - JiaXin Lu
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China
| | - YongLiang Jiang
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (The First-Affiliated Hospital of Hunan Normal University), No. 61 Jiefang Xi Road, Changsha, Hunan, 410219, China.
| |
Collapse
|
14
|
Peng Z, Luo XY, Li X, Li Y, Wu Y, Tian Y, Pan B, Petrovic A, Kosanovic D, Schermuly RT, Ruppert C, Günther A, Zhang Z, Qiu C, Li Y, Pu J, Li X, Chen AF. Cathepsin L Promotes Pulmonary Hypertension via BMPR2/GSDME-Mediated Pyroptosis. Hypertension 2024; 81:2430-2443. [PMID: 39403807 DOI: 10.1161/hypertensionaha.124.22903] [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: 02/18/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a fatal progressive disease characterized by pulmonary endothelial injury and occlusive pulmonary vascular remodeling. Lysosomal protease cathepsin L degrades essential molecules to participate in the human pathophysiological process. BMPR2 (bone morphogenetic protein type II receptor) deficiency, an important cause of PH, results from mutational inactivation or excessive lysosomal degradation and induces caspase-3-mediated cell death. Given recent evidence that pyroptosis, as a new form of programmed cell death, is induced by caspase-3-dependent GSDME (gasdermin E) cleavage, we hypothesized that cathepsin L might promote PH through BMPR2/caspase-3/GSDME axis-mediated pyroptosis. METHODS Cathepsin L expression was evaluated in the lungs and plasma of patients with pulmonary arterial hypertension. The role of cathepsin L in the progression of PH and vascular remodeling was assessed in vivo. Small interfering RNA, specific inhibitors, and lentiviruses were used to explore the mechanisms of cathepsin L on human pulmonary arterial endothelial cell dysfunction. RESULTS Cathepsin L expression is elevated in pulmonary artery endothelium from patients with idiopathic pulmonary arterial hypertension and experimental PH models. Genetic ablation of cathepsin L in PH rats relieved right ventricular systolic pressure, pulmonary vascular remodeling, and right ventricular hypertrophy, also restoring endothelial integrity. Mechanistically, cathepsin L promotes caspase-3/GSDME-mediated endothelial cell pyroptosis and represses BMPR2 signaling activity. Cathepsin L degrades BMPR2 via the lysosomal pathway, and restoring BMPR2 signaling prevents the pro-pyroptotic role of cathepsin L in PAECs and experimental PH models. CONCLUSIONS These results show for the first time that cathepsin L promotes the development of PH by degrading BMPR2 to induce caspase-3/GSDME-mediated endothelial pyroptosis.
Collapse
Affiliation(s)
- Zhouyangfan Peng
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Xue-Yang Luo
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Xinyi Li
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Yapei Li
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Health Management Medicine Center, the Third Xiangya Hospital (Yapei Li., Ying Li.), Central South University, Changsha, China
| | - Yusi Wu
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Yuyang Tian
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| | - Bingjie Pan
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Aleksandar Petrovic
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Djuro Kosanovic
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University, Russia (D.K.)
| | - Ralph Theo Schermuly
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Clemens Ruppert
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Andreas Günther
- Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Germany (A.P., R.T.S., C.R., A.G.)
| | - Zhen Zhang
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
| | - Chengfeng Qiu
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Ying Li
- Health Management Medicine Center, the Third Xiangya Hospital (Yapei Li., Ying Li.), Central South University, Changsha, China
| | - Jun Pu
- Department of Cardiology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital (J.P.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaohui Li
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Department of Pharmacology, Xiangya School of Pharmaceutical Science (Z.P., Y.W., B.P., C.Q., Xiaohui Li), Central South University, Changsha, China
| | - Alex F Chen
- The Center for Vascular Disease and Translational Medicine, the Third Xiangya Hospital (Z.P., Yapei Li., Y.W., B.P., Z.Z., C.Q., Xiaohui Li, A.F.C.), Central South University, Changsha, China
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital (XY. L, Xinyi Li, Y. T., A.F.C.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
15
|
Stump B, Waxman AB. Pulmonary Arterial Hypertension and TGF-β Superfamily Signaling: Focus on Sotatercept. BioDrugs 2024; 38:743-753. [PMID: 39292393 DOI: 10.1007/s40259-024-00680-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and progressive disease that continues to remain highly morbid despite multiple advances in medical therapies. There remains a persistent and desperate need to identify novel methods of treating and, ideally, reversing the pathologic vasculopathy that results in PAH development and progression. Sotatercept is a first-in-class fusion protein that is believed to primarily inhibit activin signaling resulting in decreased cell proliferation and differentiation, though the exact mechanism remains uncertain. Here, we review the currently available PAH therapies, data highlighting the importance of transforming growth factor-β (TGF-β) superfamily signaling in the development of PAH, and the published and on-going clinical trials evaluating sotatercept in the treatment of PAH. We will also discuss preclinical data supporting the potential use of the fusion protein KER-012 in the inhibition of aberrant TGF-β superfamily signaling to ameliorate the obstructive vasculopathy of PAH.
Collapse
|
16
|
Lin L, Lin Y, Han Z, Wang K, Zhou S, Wang Z, Wang S, Chen H. Understanding the molecular regulatory mechanisms of autophagy in lung disease pathogenesis. Front Immunol 2024; 15:1460023. [PMID: 39544928 PMCID: PMC11560454 DOI: 10.3389/fimmu.2024.1460023] [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: 07/05/2024] [Accepted: 10/07/2024] [Indexed: 11/17/2024] Open
Abstract
Lung disease development involves multiple cellular processes, including inflammation, cell death, and proliferation. Research increasingly indicates that autophagy and its regulatory proteins can influence inflammation, programmed cell death, cell proliferation, and innate immune responses. Autophagy plays a vital role in the maintenance of homeostasis and the adaptation of eukaryotic cells to stress by enabling the chelation, transport, and degradation of subcellular components, including proteins and organelles. This process is essential for sustaining cellular balance and ensuring the health of the mitochondrial population. Recent studies have begun to explore the connection between autophagy and the development of different lung diseases. This article reviews the latest findings on the molecular regulatory mechanisms of autophagy in lung diseases, with an emphasis on potential targeted therapies for autophagy.
Collapse
Affiliation(s)
- Lin Lin
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zhongyu Han
- School of Medicine, Southeast University, Nanjing, China
- Science Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Ke Wang
- Department of Science and Education, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, China
| | - Shuwei Zhou
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing, China
| | - Zhanzhan Wang
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Lianyungang, Lianyungang, China
| | - Siyu Wang
- Department of Preventive Medicine, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Haoran Chen
- Science Education Department, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
17
|
He S, Bai J, Zhang L, Yuan H, Ma C, Wang X, Guan X, Mei J, Zhu X, Xin W, Zhu D. Superenhancer-driven circRNA Myst4 involves in pulmonary artery smooth muscle cell ferroptosis in pulmonary hypertension. iScience 2024; 27:110900. [PMID: 39351203 PMCID: PMC11440257 DOI: 10.1016/j.isci.2024.110900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/19/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
The abnormal expression of circular RNAs (circRNAs) is emerging as a critical cause in regulation of pathological changes of hypoxic pulmonary hypertension (PH), in which ferroptosis is a new pathological change reported recently. However, how circRNAs regulate ferroptosis remains unclear. Here, we proved a significant decrease in circMyst4 expression in hypoxia. In vitro assays revealed that circMyst4 alleviated hypoxic pulmonary artery smooth muscle cell (PASMC) ferroptosis through directly combing with DDX5 in the nucleus to promote GPX4 mRNA processing and inhibiting the formation of the Eef1a1/ACSL4 complex in the cytoplasm. Additionally, superenhancer (SE) was verified to drive the generation of circMyst4. In vivo assays revealed that circMyst4 inhibited the progression of hypoxic PH. Overall, SE-driven circMyst4 may be a new potential therapeutic target for mediating PASMC ferroptosis through promoting DDX5-regulated GPX4 mRNA processing and inhibiting the binding between Eef1a1 and ACSL4.
Collapse
Affiliation(s)
- Siyu He
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - June Bai
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Hao Yuan
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - Cui Ma
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xiaoying Wang
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xiaoyu Guan
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
| | - Jian Mei
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xiangrui Zhu
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Wei Xin
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200031, P.R. China
| | - Daling Zhu
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
- College of Pharmacy, Harbin Medical University, Harbin 150081, P.R. China
- Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, Harbin 150081, P.R. China
| |
Collapse
|
18
|
Song Y, Jia H, Ma Q, Zhang L, Lai X, Wang Y. The causes of pulmonary hypertension and the benefits of aerobic exercise for pulmonary hypertension from an integrated perspective. Front Physiol 2024; 15:1461519. [PMID: 39483752 PMCID: PMC11525220 DOI: 10.3389/fphys.2024.1461519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 09/26/2024] [Indexed: 11/03/2024] Open
Abstract
Pulmonary hypertension is a progressive disease of the pulmonary arteries that begins with increased pulmonary artery pressure, driven by progressive remodeling of the small pulmonary arteries, and ultimately leads to right heart failure and death. Vascular remodeling is the main pathological feature of pulmonary hypertension, but treatments for pulmonary hypertension are lacking. Determining the process of vascular proliferation and dysfunction may be a way to decipher the pathogenesis of pulmonary hypertension. In this review, we summarize the important pathways of pulmonary hypertension pathogenesis. We show how these processes are integrated and emphasize the benign role of aerobic exercise, which, as an adjunctive therapy, may be able to modify vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Yinping Song
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Hao Jia
- School of Physical Education, Shaanxi Normal University, Xi’an, China
| | - Qing Ma
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Lulu Zhang
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Xiangyi Lai
- School of Physical Education, Xi’an Fanyi University, Xi’an, China
| | - Youhua Wang
- School of Physical Education, Shaanxi Normal University, Xi’an, China
| |
Collapse
|
19
|
Deliu N, Das R, May A, Newman J, Steele J, Duckworth M, Jones RJ, Wilkins MR, Toshner MR, Villar SS. StratosPHere 2: study protocol for a response-adaptive randomised placebo-controlled phase II trial to evaluate hydroxychloroquine and phenylbutyrate in pulmonary arterial hypertension caused by mutations in BMPR2. Trials 2024; 25:680. [PMID: 39407331 PMCID: PMC11475842 DOI: 10.1186/s13063-024-08485-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Pulmonary arterial hypertension is a life-threatening progressive disorder characterised by high blood pressure (hypertension) in the arteries of the lungs (pulmonary artery). Although treatable, there is no known cure for this rare disorder, and its exact cause is unknown. Mutations in the bone morphogenetic protein receptor type-2 (BMPR2) are the most common genetic cause of familial pulmonary arterial hypertension. This study represents the first-ever trial of treatments aimed at directly rescuing the BMPR2 pathway, repurposing two drugs that have shown promise at restoring levels of BMPR2 signalling: hydroxychloroquine and phenylbutyrate. METHODS This three-armed phase II precision medicine study will investigate BMPR2 target engagement and explore the efficacy of two repurposed therapies in pulmonary arterial hypertension patients with BMPR2 mutations. Patients will be stratified based on two BMPR2 mutation classes: missense and haploinsufficient mutations. Eligible subjects will be randomised to one of the three arms (two active therapy arms and a placebo arm, all plus standard of care) following a Bayesian response-adaptive design implemented independently in each stratum and updated in response to a novel panel of primary biomarkers designed to assess biological modification of the disease. DISCUSSION The results of this trial will provide the first randomised evidence of the efficacy of these therapies to rescue BMPR2 function and will efficiently explore the potential for a differential response of these therapies per mutation class to address causes rather than consequences of this rare disease. TRIAL REGISTRATION The study has been registered with ISRCTN (ISRCTN10304915, 22/09/2023).
Collapse
Affiliation(s)
- Nina Deliu
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK.
- MEMOTEF, Sapienza University, Rome, Italy.
| | - Rajenki Das
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK
| | - Angelique May
- VPD-HLRI, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Joseph Newman
- VPD-HLRI, Department of Medicine, University of Cambridge, Cambridge, UK
- Royal Papworth Hospital, Cambridge, UK
| | - Jo Steele
- Papworth Trials Unit Collaboration, Royal Papworth Hospital, Cambridge, UK
| | - Melissa Duckworth
- Papworth Trials Unit Collaboration, Royal Papworth Hospital, Cambridge, UK
| | - Rowena J Jones
- VPD-HLRI, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin R Wilkins
- Imperial College London, Heart Lung Research Institute, London, UK
| | - Mark R Toshner
- VPD-HLRI, Department of Medicine, University of Cambridge, Cambridge, UK
- Royal Papworth Hospital, Cambridge, UK
| | - Sofia S Villar
- MRC Biostatistics Unit, Cambridge University, Cambridge, UK
| |
Collapse
|
20
|
Steinhauser ML, Maron BA. Viewing Pulmonary Arterial Hypertension Pathogenesis and Opportunities for Disease-Modifying Therapy Through the Lens of Biomass. JACC Basic Transl Sci 2024; 9:1252-1263. [PMID: 39534642 PMCID: PMC11551874 DOI: 10.1016/j.jacbts.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 11/16/2024]
Abstract
Fibroproliferative remodeling of distal pulmonary arterioles is a cornerstone characteristic of pulmonary arterial hypertension (PAH). Data from contemporary quantitative imaging suggest that anabolic synthesis of macromolecular substrate, defined here as biomass, is the proximate event that causes vascular remodeling via pathogenic changes to DNA, collagen, cytoskeleton, and lipid membranes. Modifying biomass is achievable but requires tilting the balance in favor of endogenous degradation over synthetic pathways in order to advance the first-ever disease-modifying PAH pharmacotherapy. Viewing PAH pathobiology through the lens of biomass represents an opportunity to decipher novel determinants of disease inception and inform interventions that induce reverse remodeling.
Collapse
Affiliation(s)
- Matthew L. Steinhauser
- Division of Cardiovascular Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bradley A. Maron
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
- University of Maryland-Institute for Health Computing, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Wan JJ, Yi J, Wang FY, Li X, Zhang C, Song L, Dai AG. Role of mitophagy in pulmonary hypertension: Targeting the mechanism and pharmacological intervention. Mitochondrion 2024; 78:101928. [PMID: 38992857 DOI: 10.1016/j.mito.2024.101928] [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: 11/20/2023] [Revised: 05/29/2024] [Accepted: 07/07/2024] [Indexed: 07/13/2024]
Abstract
Mitophagy, a crucial pathway in eukaryotic cells, selectively eliminates dysfunctional mitochondria, thereby maintaining cellular homeostasis via mitochondrial quality control. Pulmonary hypertension (PH) refers to a pathological condition where pulmonary arterial pressure is abnormally elevated due to various reasons, and the underlying pathogenesis remains elusive. This article examines the molecular mechanisms underlying mitophagy, emphasizing its role in PH and the progress in elucidating related molecular signaling pathways. Additionally, it highlights current drug regulatory pathways, aiming to provide novel insights into the prevention and treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Jia-Jing Wan
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Jian Yi
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China
| | - Fei-Ying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Xia Li
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Chao Zhang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Lan Song
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China
| | - Ai-Guo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha 410208, Hunan, China; Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, China; Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410021, Hunan, China.
| |
Collapse
|
22
|
Zhang Q, Yaoita N, Tabuchi A, Liu S, Chen SH, Li Q, Hegemann N, Li C, Rodor J, Timm S, Laban H, Finkel T, Stevens T, Alvarez DF, Erfinanda L, de Perrot M, Kucherenko MM, Knosalla C, Ochs M, Dimmeler S, Korff T, Verma S, Baker AH, Kuebler WM. Endothelial Heterogeneity in the Response to Autophagy Drives Small Vessel Muscularization in Pulmonary Hypertension. Circulation 2024; 150:466-487. [PMID: 38873770 DOI: 10.1161/circulationaha.124.068726] [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: 01/26/2024] [Accepted: 04/18/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Endothelial cell (EC) apoptosis and proliferation of apoptosis-resistant cells is a hallmark of pulmonary hypertension (PH). Yet, why some ECs die and others proliferate and how this contributes to vascular remodeling is unclear. We hypothesized that this differential response may: (1) relate to different EC subsets, namely pulmonary artery (PAECs) versus microvascular ECs (MVECs); (2) be attributable to autophagic activation in both EC subtypes; and (3) cause replacement of MVECs by PAECs with subsequent distal vessel muscularization. METHODS EC subset responses to chronic hypoxia were assessed by single-cell RNA sequencing of murine lungs. Proliferative versus apoptotic responses, activation, and role of autophagy were assessed in human and rat PAECs and MVECs, and in precision-cut lung slices of wild-type mice or mice with endothelial deficiency in the autophagy-related gene 7 (Atg7EN-KO). Abundance of PAECs versus MVECs in precapillary microvessels was assessed in lung tissue from patients with PH and animal models on the basis of structural or surface markers. RESULTS In vitro and in vivo, PAECs proliferated in response to hypoxia, whereas MVECs underwent apoptosis. Single-cell RNA sequencing analyses support these findings in that hypoxia induced an antiapoptotic, proliferative phenotype in arterial ECs, whereas capillary ECs showed a propensity for cell death. These distinct responses were prevented in hypoxic Atg7EN-KO mice or after ATG7 silencing, yet replicated by autophagy stimulation. In lung tissue from mice, rats, or patients with PH, the abundance of PAECs in precapillary arterioles was increased, and that of MVECs reduced relative to controls, indicating replacement of microvascular by macrovascular ECs. EC replacement was prevented by genetic or pharmacological inhibition of autophagy in vivo. Conditioned medium from hypoxic PAECs yet not MVECs promoted pulmonary artery smooth muscle cell proliferation and migration in a platelet-derived growth factor-dependent manner. Autophagy inhibition attenuated PH development and distal vessel muscularization in preclinical models. CONCLUSIONS Autophagic activation by hypoxia induces in parallel PAEC proliferation and MVEC apoptosis. These differential responses cause a progressive replacement of MVECs by PAECs in precapillary pulmonary arterioles, thus providing a macrovascular context that in turn promotes pulmonary artery smooth muscle cell proliferation and migration, ultimately driving distal vessel muscularization and the development of PH.
Collapse
Affiliation(s)
- Qi Zhang
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- Department of Cardiology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q.Z.)
| | - Nobuhiro Yaoita
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Arata Tabuchi
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Shaofei Liu
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
| | - Shiau-Haln Chen
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (S.-H.C., J.R., A.H.B.)
| | - Qiuhua Li
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Niklas Hegemann
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany (N.H., M.M.K., C.K.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Germany (N.H., M.M.K., C.K.)
| | - Caihong Li
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Julie Rodor
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (S.-H.C., J.R., A.H.B.)
| | - Sara Timm
- Core Facility Electron Microscopy (S.T., M.O.), Charité-Universitätsmedizin, Berlin, Germany
| | - Hebatullah Laban
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology (H.L.), Heidelberg University, Germany
- German Center for Cardiovascular Research, Partner Site Heidelberg/Mannheim, Heidelberg (H.L.)
| | - Toren Finkel
- Department of Medicine, Division of Cardiology, University of Pittsburgh, PA (T.F.)
| | - Troy Stevens
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile (T.S.)
| | - Diego F Alvarez
- Department of Physiology and Pharmacology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX (D.F.A.)
| | - Lasti Erfinanda
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
| | - Marc de Perrot
- Division of Thoracic Surgery, Toronto General Hospital, Canada (M.d.P.)
- Department of Surgery (M.d.P., W.M.K.), University of Toronto, Canada
| | - Mariya M Kucherenko
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany (N.H., M.M.K., C.K.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Germany (N.H., M.M.K., C.K.)
| | - Christoph Knosalla
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité, Berlin, Germany (N.H., M.M.K., C.K.)
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Germany (N.H., M.M.K., C.K.)
| | - Matthias Ochs
- Core Facility Electron Microscopy (S.T., M.O.), Charité-Universitätsmedizin, Berlin, Germany
- Institute of Functional Anatomy (M.O.), Charité-Universitätsmedizin, Berlin, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany (S.D.)
| | - Thomas Korff
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology (T.K.), Heidelberg University, Germany
- European Center for Angioscience, Medical Faculty Mannheim (T.K.), Heidelberg University, Germany
| | - Subodh Verma
- Division of Cardiac Surgery (S.V.), University of Toronto, Canada
| | - Andrew H Baker
- Centre for Cardiovascular Science, University of Edinburgh, United Kingdom (S.-H.C., J.R., A.H.B.)
- Department of Pathology, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases, Maastricht University, The Netherlands (A.H.B.)
| | - Wolfgang M Kuebler
- Institute of Physiology (Q.Z., N.Y., A.T., S.L., Q.L., N.H., C.L., L.E., M.M.K., W.M.K.), Charité-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research, Partner Site Berlin (S.L., N.H., M.M.K., C.K., W.M.K.)
- Department of Surgery (M.d.P., W.M.K.), University of Toronto, Canada
- Department of Physiology (W.M.K.), University of Toronto, Canada
- Keenan Research Centre, St Michael's Hospital, Canada (W.M.K.)
| |
Collapse
|
23
|
Fareed SA, Yousef EM, Abd El-Moneam SM. Effects of prolonged hydroxychloroquine use on the pancreatic tissue and expected ameliorative effect of lactoferrin in rats (biochemical, histological, and morphometric study). Tissue Cell 2024; 89:102439. [PMID: 38889555 DOI: 10.1016/j.tice.2024.102439] [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: 01/07/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024]
Abstract
Hydroxychloroquine (HCQ), an antimalarial drug widely used in treating rheumatoid disorders. Many side effects have been reported with HCQ administration indicating its hazardous effects on various organs. No previous studies reported the effect of long-term administration of oral HCQ on pancreatic tissue. Our study assessed pancreatic tissues functional and histopathological alterations following prolonged oral administration of HCQ. We also investigated the possible ameliorative effects of the lactoferrin (LF) coadministration with HCQ in adult male albino rats. Forty adult male Wister albino rats were divided into: negative control, LF positive control (2 g/kg), HCQ-treated (200 mg/kg), and HCQ+LF treated. Biochemical, histological, immunohistochemical, and morphometric analyses of the pancreatic tissues were conducted. Our findings revealed that prolonged oral administration of HCQ induced significant disruption of the pancreatic acinar architecture, enlarged congested islets of Langerhans, and elevated plasma insulin, amylase, and lipase levels. Interestingly, LF administration ameliorated the deleterious effects of prolonged HCQ administration on pancreatic tissue of adult male albino rats. In conclusion, prolonged oral administration of HCQ induced pancreatic tissue damage in rats, while LF attenuates HCQ-induced pancreatic injury. Our results emphasized the necessity of prescribing HCQ with caution, considering both dosage and treatment duration.
Collapse
Affiliation(s)
- Shimaa Antar Fareed
- Department of Human Anatomy & Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| | - Einas Mohamed Yousef
- Department of Histology and Cell Biology, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| | - Samar M Abd El-Moneam
- Department of Human Anatomy & Embryology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
24
|
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
|
25
|
Saddouk FZ, Kuzemczak A, Saito J, Greif DM. Endothelial HIFα/PDGF-B to smooth muscle Beclin1 signaling sustains pathological muscularization in pulmonary hypertension. JCI Insight 2024; 9:e162449. [PMID: 38652543 PMCID: PMC11141934 DOI: 10.1172/jci.insight.162449] [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: 06/07/2022] [Accepted: 04/17/2024] [Indexed: 04/25/2024] Open
Abstract
Mechanisms underlying maintenance of pathological vascular hypermuscularization are poorly delineated. Herein, we investigated retention of smooth muscle cells (SMCs) coating normally unmuscularized distal pulmonary arterioles in pulmonary hypertension (PH) mediated by chronic hypoxia with or without Sugen 5416, and reversal of this pathology. With hypoxia in mice or culture, lung endothelial cells (ECs) upregulated hypoxia-inducible factor 1α (HIF1-α) and HIF2-α, which induce platelet-derived growth factor B (PDGF-B), and these factors were reduced to normoxic levels with re-normoxia. Re-normoxia reversed hypoxia-induced pulmonary vascular remodeling, but with EC HIFα overexpression during re-normoxia, pathological changes persisted. Conversely, after establishment of distal muscularization and PH, EC-specific deletion of Hif1a, Hif2a, or Pdgfb induced reversal. In human idiopathic pulmonary artery hypertension, HIF1-α, HIF2-α, PDGF-B, and autophagy-mediating gene products, including Beclin1, were upregulated in pulmonary artery SMCs and/or lung lysates. Furthermore, in mice, hypoxia-induced EC-derived PDGF-B upregulated Beclin1 in distal arteriole SMCs, and after distal muscularization was established, re-normoxia, EC Pdgfb deletion, or treatment with STI571 (which inhibits PDGF receptors) downregulated SMC Beclin1 and other autophagy products. Finally, SMC-specific Becn1 deletion induced apoptosis, reversing distal muscularization and PH mediated by hypoxia with or without Sugen 5416. Thus, chronic hypoxia induction of the HIFα/PDGF-B axis in ECs is required for non-cell-autonomous Beclin1-mediated survival of pathological distal arteriole SMCs.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Arterioles/metabolism
- Arterioles/pathology
- Autophagy
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Beclin-1/metabolism
- Beclin-1/genetics
- Disease Models, Animal
- Endothelial Cells/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Hypoxia/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Indoles
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Proto-Oncogene Proteins c-sis/metabolism
- Proto-Oncogene Proteins c-sis/genetics
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pyrroles
- Signal Transduction
- Vascular Remodeling
Collapse
Affiliation(s)
- Fatima Z. Saddouk
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Andrew Kuzemczak
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Junichi Saito
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, and
- Department of Genetics, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
26
|
Jiang G, Shi LF, Li LJ, Duan XJ, Zheng ZF. Activation of the p62-Keap1-Nrf2 pathway improves pulmonary arterial hypertension in MCT-induced rats by inhibiting autophagy. FASEB J 2024; 38:e23452. [PMID: 38308640 DOI: 10.1096/fj.202301563r] [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: 08/01/2023] [Revised: 11/30/2023] [Accepted: 01/12/2024] [Indexed: 02/05/2024]
Abstract
Autophagy is implicated in the pathogenesis of pulmonary arterial hypertension (PAH). We aimed to investigate whether the p62-Keap1-Nrf2 pathway affects the development of PAH by mediating autophagy. A PAH rat model was established using monocrotaline (MCT). Pulmonary artery smooth muscle cells (PASMCs) were extracted, and the changes in proliferation, migration, autophagy, and oxidative stress were analyzed following overexpression or knockdown of p62. The impact of p62 on the symptoms of PAH rats was assessed by the injection of an adenovirus overexpressing p62. We found that the knockdown of p62 increased the proliferation and migration of PASMCs, elevating the oxidative stress of PASMCs and upregulating gene expression of NADPH oxidases. Co-IP assay results demonstrated that p62 interacted with Keap1. p62 knockdown enhanced Keap1 protein stability and Nrf2 ubiquitination. LC3II/I and ATG5 were expressed more often when p62 was knocked down. Treating with an inhibitor of autophagy reversed the impact of p62 knockdown on PASMCs. Nrf2 inhibitor treatment reduced the expression of Nrf2 and p62, while increasing the expression of Keap1, LC3II/I, and ATG5 in PASMCs. However, overexpressing p62 diminished mRVP, SPAP, and Fulton index in PAH rats and attenuated pulmonary vascular wall thickening. Overexpression of p62 also decreased the expression of Keap1, LC3II/I, and ATG5 and increased the nuclear expression of Nrf2 in PAH rats. Importantly, overexpression of p62 reduced oxidative stress and the NADPH oxidase expression in PAH rats. Overall, activation of the p62-Keap1-Nrf2 positive feedback signaling axis reduces the proliferation and migration of PASMCs and alleviates PAH by inhibiting autophagy and oxidative stress.
Collapse
Affiliation(s)
- Gang Jiang
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Li-Fang Shi
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Ling-Jiao Li
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xiao-Ju Duan
- Department of Respiratory Medicine, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Zhao-Fen Zheng
- Department of Cardiovascular Medicine, Hunan Provincial People's Hospital (the First Affiliated Hospital of Hunan Normal University), Changsha, China
| |
Collapse
|
27
|
Samare-Najaf M, Neisy A, Samareh A, Moghadam D, Jamali N, Zarei R, Zal F. The constructive and destructive impact of autophagy on both genders' reproducibility, a comprehensive review. Autophagy 2023; 19:3033-3061. [PMID: 37505071 PMCID: PMC10621263 DOI: 10.1080/15548627.2023.2238577] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Reproduction is characterized by a series of massive renovations at molecular, cellular, and tissue levels. Recent studies have strongly tended to reveal the involvement of basic molecular pathways such as autophagy, a highly conserved eukaryotic cellular recycling, during reproductive processes. This review comprehensively describes the current knowledge, updated to September 2022, of autophagy contribution during reproductive processes in males including spermatogenesis, sperm motility and viability, and male sex hormones and females including germ cells and oocytes viability, ovulation, implantation, fertilization, and female sex hormones. Furthermore, the consequences of disruption in autophagic flux on the reproductive disorders including oligospermia, azoospermia, asthenozoospermia, teratozoospermia, globozoospermia, premature ovarian insufficiency, polycystic ovarian syndrome, endometriosis, and other disorders related to infertility are discussed as well.Abbreviations: AKT/protein kinase B: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; E2: estrogen; EDs: endocrine disruptors; ER: endoplasmic reticulum; FSH: follicle stimulating hormone; FOX: forkhead box; GCs: granulosa cells; HIF: hypoxia inducible factor; IVF: in vitro fertilization; IVM: in vitro maturation; LCs: Leydig cells; LDs: lipid droplets; LH: luteinizing hormone; LRWD1: leucine rich repeats and WD repeat domain containing 1; MAP1LC3: microtubule associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; MTOR: mechanistic target of rapamycin kinase; NFKB/NF-kB: nuclear factor kappa B; P4: progesterone; PCOS: polycystic ovarian syndrome; PDLIM1: PDZ and LIM domain 1; PI3K: phosphoinositide 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns3K: class III phosphatidylinositol 3-kinase; POI: premature ovarian insufficiency; ROS: reactive oxygen species; SCs: Sertoli cells; SQSTM1/p62: sequestosome 1; TSGA10: testis specific 10; TST: testosterone; VCP: vasolin containing protein.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | - Asma Neisy
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Delaram Moghadam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Jamali
- Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Reza Zarei
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Zal
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
28
|
Luo Y, Tan Z, Ye Y, Ma X, Yue G. Qiqilian ameliorates vascular endothelial dysfunction by inhibiting NLRP3-ASC inflammasome activation in vivo and in vitro. PHARMACEUTICAL BIOLOGY 2023; 61:815-824. [PMID: 37194678 PMCID: PMC10599261 DOI: 10.1080/13880209.2023.2208617] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/25/2023] [Accepted: 04/25/2023] [Indexed: 05/18/2023]
Abstract
CONTEXT Previous studies have highlighted significant therapeutic effects of Qiqilian (QQL) capsule on hypertension in spontaneously hypertensive rats (SHRs); however, its underlying molecular mechanism remains unclear. OBEJECTIVE We investigated the potential mechanism by which QQL improves hypertension-induced vascular endothelial dysfunction (VED). MATERIALS AND METHODS In vivo, SHRs were divided into four groups (20 per group) and were administered gradient doses of QQL (0, 0.3, 0.6, and 1.2 g/kg) for 8 weeks, while Wistar Kyoto rats were used as normal control. The vascular injury extent, IL-1β and IL-18 levels, NLRP3, ASC and caspase-1 contents were examined. In vitro, the effects of QQL-medicated serum on angiotensin II (AngII)-induced inflammatory and autophagy in human umbilical vein endothelial cells (HUVECs) were assessed. RESULT Compared with the SHR group, QQL significantly decreased thickness (125.50 to 105.45 μm) and collagen density (8.61 to 3.20%) of arterial vessels, and reduced serum IL-1β (96.25 to 46.13 pg/mL) and IL-18 (345.01 to 162.63 pg/mL) levels. The NLRP3 and ACS expression in arterial vessels were downregulated (0.21- and 0.16-fold, respectively) in the QQL-HD group compared with the SHR group. In vitro, QQL treatment restored NLRP3 and ASC expression, which was downregulated approximately 2-fold compared with that of AngII-induced HUVECs. Furthermore, QQL decreased LC3II and increased p62 contents (p < 0.05), indicating a reduction in autophagosome accumulation. These effects were inhibited by the autophagy agonist rapamycin and enhanced by the autophagy inhibitor chloroquine. CONCLUSION QQL effectively attenuated endothelial injury and inflammation by inhibiting AngII-induced excessive autophagy, which serves as a potential therapeutic strategy for hypertension.
Collapse
Affiliation(s)
- Yuan Luo
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, P.R. China
| | | | - Yun Ye
- No. 923 Hospital of the PLA Joint Logistics Support Force, Nanning, P.R. China
| | - Xiaocong Ma
- Guangxi University of Chinese Medicine, Nanning, P.R. China
| | - Guihua Yue
- Guangxi Internation Zhuang Medicine Hospital to Guangxi University of Chinese Medicine, Nanning, P.R. China
- Guangxi Internation Zhuang Medicine Hospital, Nanning, P.R. China
| |
Collapse
|
29
|
Ren H, Dai R, Nik Nabil WN, Xi Z, Wang F, Xu H. Unveiling the dual role of autophagy in vascular remodelling and its related diseases. Biomed Pharmacother 2023; 168:115643. [PMID: 37839111 DOI: 10.1016/j.biopha.2023.115643] [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: 07/29/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
Vascular remodelling is an adaptive response to physiological and pathological stimuli that leads to structural and functional changes in the vascular intima, media, and adventitia. Pathological vascular remodelling is a hallmark feature of numerous vascular diseases, including atherosclerosis, hypertension, abdominal aortic aneurysm, pulmonary hypertension and preeclampsia. Autophagy is critical in maintaining cellular homeostasis, and its dysregulation has been implicated in the pathogenesis of various diseases, including vascular diseases. However, despite emerging evidence, the role of autophagy and its dual effects on vascular remodelling has garnered limited attention. Autophagy can exert protective and detrimental effects on the vascular intima, media and adventitia, thereby substantially influencing the course of vascular remodelling and its related vascular diseases. Currently, there has not been a review that thoroughly describes the regulation of autophagy in vascular remodelling and its impact on related diseases. Therefore, this review aimed to bridge this gap by focusing on the regulatory roles of autophagy in diseases related to vascular remodelling. This review also summarizes recent advancements in therapeutic agents targeting autophagy to regulate vascular remodelling. Additionally, this review offers an overview of recent breakthroughs in therapeutic agents targeting autophagy to regulate vascular remodelling. A deeper understanding of how autophagy orchestrates vascular remodelling can drive the development of targeted therapies for vascular diseases.
Collapse
Affiliation(s)
- Hangui Ren
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China; Pharmaceutical Services Program, Ministry of Health, Selangor 46200, Malaysia
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Feng Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China.
| |
Collapse
|
30
|
Prabhakar A, Kumar R, Wadhwa M, Ghatpande P, Zhang J, Zhao Z, Lizama CO, Kharbikar BN, Gräf S, Treacy CM, Morrell NW, Graham BB, Lagna G, Hata A. Reversal of pulmonary veno-occlusive disease phenotypes by inhibition of the integrated stress response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.27.568924. [PMID: 38076809 PMCID: PMC10705277 DOI: 10.1101/2023.11.27.568924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Pulmonary veno-occlusive disease (PVOD) is a rare form of pulmonary hypertension arising from EIF2AK4 gene mutations or mitomycin C (MMC) administration. The lack of effective PVOD therapies is compounded by a limited understanding of the mechanisms driving the vascular remodeling in PVOD. We show that the administration of MMC in rats mediates the activation of protein kinase R (PKR) and the integrated stress response (ISR), which lead to the release of the endothelial adhesion molecule VE-Cadherin in the complex with Rad51 to the circulation, disruption of endothelial barrier, and vascular remodeling. Pharmacological inhibition of PKR or ISR attenuates the depletion of VE-Cadherin, elevation of vascular permeability, and vascular remodeling instigated by MMC, suggesting potential clinical intervention for PVOD. Finally, the severity of PVOD phenotypes was increased by a heterozygous BMPR2 mutation that truncates the carboxyl tail of BMPR2, underscoring the role of deregulated BMP signal in the development of PVOD.
Collapse
|
31
|
Xu L, Qu C, Liu Y, Liu H. The environmental enrichment ameliorates chronic cerebral hypoperfusion-induced cognitive impairment by activating autophagy signaling pathway and improving synaptic function in hippocampus. Brain Res Bull 2023; 204:110798. [PMID: 37890595 DOI: 10.1016/j.brainresbull.2023.110798] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 10/01/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a frequently observed underlying pathology of both Alzheimer's disease (AD) and vascular dementia (VD), which is a common consequence of cerebral blood flow (CBF) dysregulation. Synaptic damage has been proven as a crucial causative factor for CCH-related cognitive impairment. This study aimed to investigate the neuroprotective impact of environmental enrichment (EE) intervention on CCH-induced synaptic destruction and the consequent cognitive impairment. Furthermore, the underlying mechanism of this neuroprotective effect was explored to provide new insights into therapeutic interventions for individuals suffering from AD or VD. METHODS In this experiment, all rats were initially acclimatized to a standard environment (SE) for a period of one week. On the seventh day, rats underwent either bilateral common carotid artery occlusion (2VO) surgery or sham surgery (Sham) before being subjected to a four-week procedure of exposure to an EE, except for the control group. During the EE or SE procedure, intraperitoneal injection of chloroquine (CQ) into rats was performed once daily for four weeks. Following this, cognitive function was assessed using the Morris water maze (MWM) test. The synapse ultrastructure was subsequently observed using transmission electron microscopy. Expression levels of autophagy-related proteins (LC3, LAMP1, and P62) and synapse-related proteins (Synapsin I and PSD-95) were detected through Western blotting. Finally, immunofluorescence was used to examine the expression levels of Synapsin I and PSD-95 and the colocalization of LAMP-1 and LC3 in the hippocampus. RESULTS After undergoing 2VO, rats exposed to SE exhibited cognitive impairment, autophagic dysfunction, and synapse damage. The synapse damage was evidenced by ultrastructural damage and degradation of synapse-related proteins. However, these effects were significantly mitigated by exposure to an EE intervention. Moreover, the intervention led to an improvement in autophagic dysfunction. CONCLUSION The study found that EE had a positive impact on CCH-induced synaptic damage. Specifically, EE was found to increase synaptic plasticity-associated proteins and postsynaptic density thickness, while decreasing synaptic space. This multifaceted effect resulted in an amelioration of CCH-induced cognitive impairment. It was shown that this beneficial outcome was mediated via the activation of the autophagy-lysosomal pathway. Overall, the findings suggest that EE may have a therapeutic potential for cognitive impairments associated with CCH through autophagy-mediated synaptic improvement.
Collapse
Affiliation(s)
- Linling Xu
- The Affiliated Hospital of Southwest Jiaotong University & the Third People's Hospital of Chengdu, No.82, Qinglong Road, Chengdu 610014, Sichuan, China; Department of Neurology, Zhongnan Hospital, Wuhan University, No.169, Donghu Road, Wuhan 430071, Hubei, China
| | - Changhua Qu
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Disease, Department of Neurology, Minda Hospital of Hubei Minzu University, Enshi, China
| | - Yan Liu
- The Affiliated Hospital of Southwest Jiaotong University & the Third People's Hospital of Chengdu, No.82, Qinglong Road, Chengdu 610014, Sichuan, China
| | - Hua Liu
- The Affiliated Hospital of Southwest Jiaotong University & the Third People's Hospital of Chengdu, No.82, Qinglong Road, Chengdu 610014, Sichuan, China.
| |
Collapse
|
32
|
Chai L, Wang Q, Wang Y, Li D, Zhang Q, Chen Y, Liu J, Chen H, Qiu Y, Shen N, Wang J, Xie X, Li M. Downregulation of PDCD4 through STAT3/ATF6/autophagy mediates MIF-induced PASMCs proliferation/migration and vascular remodeling. Eur J Pharmacol 2023; 956:175968. [PMID: 37549728 DOI: 10.1016/j.ejphar.2023.175968] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 07/08/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
To address the molecular mechanisms underlying macrophage migration inhibitory factor (MIF) induced pulmonary artery smooth muscle cells (PASMCs) proliferation, migration and vascular remodeling in pulmonary hypertension (PH), primary cultured rat PASMCs and monocrotaline (MCT)-induced rats with PH were applied in the present study. The results showed that MIF increased signal transducer and activator of transcription 3 (STAT3) phosphorylation, and then stimulated activating transcription factor 6 (ATF6) activation, subsequently triggered autophagy activation, which further led to programmed cell death factor 4 (PDCD4) lysosomal degradation, and eventually promoted PASMCs proliferation/migration. In lung tissues of MCT rats, MIF protein expression was elevated, phosphorylation of STAT3 and activation of ATF6 were increased, activation of autophagy was evident, and reduction of PDCD4 was observed. Intervention with MIF inhibitor 4-Iodo-6-phenylpyrimidine (4-IPP), ATF6 blocker melatonin or autophagy inhibitor chloroquine, confirmed the in vitro interaction among MIF, STAT3, ATF6, autophagy and PDCD4 in MCT induced rats with PH. Targeting MIF/STAT3/ATF6/autophagy/PDCD4 axis effectively prevented the development of PH by suppressing PASMCs proliferation and vascular remodeling. In conclusions, we demonstrate that MIF activates the STAT3/ATF6/autophagy cascade and then degrades PDCD4 leading to PASMCs proliferation/migration and pulmonary vascular remodeling, suggesting that intervention this axis might have potential value in management of PH.
Collapse
Affiliation(s)
- Limin Chai
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qingting Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yan Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Danyang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yuqian Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jin Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Huan Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Yuanjie Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Nirui Shen
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Jian Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Xinming Xie
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China
| | - Manxiang Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Xi'an JiaoTong, University, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
33
|
Abstract
Advanced glycation end products (AGEs), by-products of glucose metabolism, have been linked to the emergence of cardiovascular disorders (CVD). AGEs can cause tissue damage in four different ways: (1) by altering protein function, (2) by crosslinking proteins, which makes tissue stiffer, (3) by causing the generation of free radicals, and (4) by activating an inflammatory response after binding particular AGE receptors, such as the receptor for advanced glycation end products (RAGE). It is suggested that the soluble form of RAGE (sRAGE) blocks ligand-mediated pro-inflammatory and oxidant activities by serving as a decoy. Therefore, several studies have investigated the possible anti-inflammatory and anti-oxidant characteristics of sRAGE, which may help lower the risk of CVD. According to the results of various studies, the relationship between circulating sRAGE, cRAGE, and esRAGE and CVD is inconsistent. To establish the potential function of sRAGE as a therapeutic target in the treatment of cardiovascular illnesses, additional studies are required to better understand the relationship between sRAGE and CVD. In this review, we explored the potential function of sRAGE in different CVD, highlighting unanswered concerns and outlining the possibilities for further investigation.
Collapse
Affiliation(s)
- Charlotte Delrue
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium
| | - Joris R Delanghe
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Marijn M Speeckaert
- Department of Nephrology, Ghent University Hospital, Ghent, Belgium; Research Foundation-Flanders (FWO), Brussels, Belgium.
| |
Collapse
|
34
|
Zhao Y, Wu J, Guan S, Xue T, Wei X, Cao D, Kong P, Zhang X. PIF1 Promotes Autophagy to Inhibit Chronic Hypoxia Induced Apoptosis of Pulmonary Artery Endothelial Cells. Int J Chron Obstruct Pulmon Dis 2023; 18:1319-1332. [PMID: 37396201 PMCID: PMC10312211 DOI: 10.2147/copd.s406453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/14/2023] [Indexed: 07/04/2023] Open
Abstract
Purpose Pulmonary artery hypertension (PAH) is a common complication of chronic obstructive pulmonary disease and obstructive sleep apnea/hypopnea syndrome worldwide. Pulmonary vascular alterations associated with PAH have multifactorial causes, in which endothelial cells play an important role. Autophagy is closely related to endothelial cell injury and the development of PAH. PIF1 is a multifunctional helicase crucial for cell survival. The present study investigated the effect of PIF1 on autophagy and apoptosis in human pulmonary artery endothelial cells (HPAECs) under chronic hypoxia stress. Methods Chronic hypoxia Gene expression profiling chip-assays identified the PIF1 gene as differentially expressed, which was verified by RT-qPCR analysis. Electron microscopy, immunofluorescence, and Western blotting were used to analyze autophagy and the expression of LC3 and P62. Apoptosis was analyzed using flow cytometry. Results Our study found that chronic hypoxia induces autophagy in HPAECs, and apoptosis was exacerbated by inhibiting autophagy. Levels of the DNA helicase PIF1 were increased in HPAECs after chronic hypoxia. PIF1 knockdown inhibited autophagy and promoted the apoptosis of HPAECs under chronic hypoxia stress. Conclusion Based on these findings, we conclude that PIF1 inhibits the apoptosis of HPAECs by accelerating the autophagy pathway. Therefore, PIF1 plays a crucial role in HPAEC dysfunction in chronic hypoxia-induced PAH and may be a potential target for the treatment of PAH.
Collapse
Affiliation(s)
- Yujing Zhao
- Department of the First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Juan Wu
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Shuai Guan
- Department of Pediatric Infectious Diseases, The First People’s Hospital of Datong, Datong, Shanxi, People’s Republic of China
| | - Ting Xue
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xiaolei Wei
- Department of the First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Dawei Cao
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Pengzhou Kong
- Department of Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| | - Xinri Zhang
- Department of NHC Key Laboratory of Pneumoconiosis, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Shanxi Key Laboratory of Respiratory Diseases, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, People’s Republic of China
| |
Collapse
|
35
|
Mao M, Song S, Li X, Lu J, Li J, Zhao W, Liu H, Liu J, Zeng B. Advances in epigenetic modifications of autophagic process in pulmonary hypertension. Front Immunol 2023; 14:1206406. [PMID: 37398657 PMCID: PMC10313199 DOI: 10.3389/fimmu.2023.1206406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Pulmonary hypertension is characterized by pulmonary arterial remodeling that results in increased pulmonary vascular resistance, right ventricular failure, and premature death. It is a threat to public health globally. Autophagy, as a highly conserved self-digestion process, plays crucial roles with autophagy-related (ATG) proteins in various diseases. The components of autophagy in the cytoplasm have been studied for decades and multiple studies have provided evidence of the importance of autophagic dysfunction in pulmonary hypertension. The status of autophagy plays a dynamic suppressive or promotive role in different contexts and stages of pulmonary hypertension development. Although the components of autophagy have been well studied, the molecular basis for the epigenetic regulation of autophagy is less understood and has drawn increasing attention in recent years. Epigenetic mechanisms include histone modifications, chromatin modifications, DNA methylation, RNA alternative splicing, and non-coding RNAs, which control gene activity and the development of an organism. In this review, we summarize the current research progress on epigenetic modifications in the autophagic process, which have the potential to be crucial and powerful therapeutic targets against the autophagic process in pulmonary hypertension development.
Collapse
Affiliation(s)
- Min Mao
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- National Health Commission (NHC) Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Xin Li
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- National Health Commission (NHC) Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jiayao Lu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Jie Li
- Marketing Department, Shenzhen Reyson Biotechnology Co., Ltd, Shenzhen, China
- Nanjing Evertop Electronics Ltd., Nanjing, China
| | - Weifang Zhao
- Quality Management Department International Registration, North China Pharmaceutical Co., Ltd. (NCPC), Hebei Huamin Pharmaceutical Co., Ltd., Shijiazhuang, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- National Health Commission (NHC) Key Laboratory of Chronobiology (Sichuan University), Chengdu, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children’s Health, West China Second University Hospital, Sichuan University, Chengdu, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jingxin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Bin Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
36
|
Wu JH, Lee JC, Ho CC, Chiu PW, Sun CH. A myeloid leukemia factor homolog is involved in tolerance to stresses and stress-induced protein metabolism in Giardia lamblia. Biol Direct 2023; 18:20. [PMID: 37095576 PMCID: PMC10127389 DOI: 10.1186/s13062-023-00378-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/17/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND The eukaryotic membrane vesicles contain specific sets of proteins that determine vesicle function and shuttle with specific destination. Giardia lamblia contains unknown cytosolic vesicles that are related to the identification of a homolog of human myeloid leukemia factor (MLF) named MLF vesicles (MLFVs). Previous studies suggest that MLF also colocalized with two autophagy machineries, FYVE and ATG8-like protein, and that MLFVs are stress-induced compartments for substrates of the proteasome or autophagy in response to rapamycin, MG132, and chloroquine treatment. A mutant protein of cyclin-dependent kinase 2, CDK2m3, was used to understand whether the aberrant proteins are targeted to degradative compratments. Interestingly, MLF was upregulated by CDK2m3 and they both colocalized within the same vesicles. Autophagy is a self-digestion process that is activated to remove damaged proteins for preventing cell death in response to various stresses. Because of the absence of some autophagy machineries, the mechanism of autophagy is unclear in G. lamblia. RESULTS In this study, we tested the six autophagosome and stress inducers in mammalian cells, including MG132, rapamycin, chloroquine, nocodazole, DTT, and G418, and found that their treatment increased reactive oxygen species production and vesicle number and level of MLF, FYVE, and ATG8-like protein in G. lamblia. Five stress inducers also increased the CDK2m3 protein levels and vesicles. Using stress inducers and knockdown system for MLF, we identified that stress induction of CDK2m3 was positively regulated by MLF. An autophagosome-reducing agent, 3-methyl adenine, can reduce MLF and CDK2m3 vesicles and proteins. In addition, knockdown of MLF with CRISPR/Cas9 system reduced cell survival upon treatment with stress inducers. Our newly developed complementation system for CRISPR/Cas9 indicated that complementation of MLF restored cell survival in response to stress inducers. Furthermore, human MLF2, like Giardia MLF, can increase cyst wall protein expression and cyst formation in G. lamblia, and it can colocalize with MLFVs and interact with MLF. CONCLUSIONS Our results suggest that MLF family proteins are functionally conserved in evolution. Our results also suggest an important role of MLF in survival in stress conditions and that MLFVs share similar stress-induced characteristics with autophagy compartments.
Collapse
Affiliation(s)
- Jui-Hsuan Wu
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan, Republic of China
| | - Jen-Chi Lee
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan, Republic of China
| | - Chun-Che Ho
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan, Republic of China
| | - Pei-Wei Chiu
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan, Republic of China
| | - Chin-Hung Sun
- Department of Tropical Medicine and Parasitology, College of Medicine, National Taiwan University, Taipei, 100, Taiwan, Republic of China.
| |
Collapse
|
37
|
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
|
38
|
Yang L, Wan N, Gong F, Wang X, Feng L, Liu G. Transcription factors and potential therapeutic targets for pulmonary hypertension. Front Cell Dev Biol 2023; 11:1132060. [PMID: 37009479 PMCID: PMC10064017 DOI: 10.3389/fcell.2023.1132060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Pulmonary hypertension (PH) is a refractory and fatal disease characterized by excessive pulmonary arterial cell remodeling. Uncontrolled proliferation and hypertrophy of pulmonary arterial smooth muscle cells (PASMCs), dysfunction of pulmonary arterial endothelial cells (PAECs), and abnormal perivascular infiltration of immune cells result in pulmonary arterial remodeling, followed by increased pulmonary vascular resistance and pulmonary pressure. Although various drugs targeting nitric oxide, endothelin-1 and prostacyclin pathways have been used in clinical settings, the mortality of pulmonary hypertension remains high. Multiple molecular abnormalities have been implicated in pulmonary hypertension, changes in numerous transcription factors have been identified as key regulators in pulmonary hypertension, and a role for pulmonary vascular remodeling has been highlighted. This review consolidates evidence linking transcription factors and their molecular mechanisms, from pulmonary vascular intima PAECs, vascular media PASMCs, and pulmonary arterial adventitia fibroblasts to pulmonary inflammatory cells. These findings will improve the understanding of particularly interactions between transcription factor-mediated cellular signaling pathways and identify novel therapies for pulmonary hypertension.
Collapse
Affiliation(s)
- Liu Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Naifu Wan
- Department of Vascular & Cardiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fanpeng Gong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xianfeng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Guizhu Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Guizhu Liu,
| |
Collapse
|
39
|
Kukreja RC, Wang R, Koka S, Das A, Samidurai A, Xi L. Treating diabetes with combination of phosphodiesterase 5 inhibitors and hydroxychloroquine-a possible prevention strategy for COVID-19? Mol Cell Biochem 2023; 478:679-696. [PMID: 36036333 PMCID: PMC9421626 DOI: 10.1007/s11010-022-04520-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/30/2022] [Indexed: 01/09/2023]
Abstract
Type 2 diabetes (T2D) is one of the major risk factors for developing cardiovascular disease and the resultant devastating morbidity and mortality. The key features of T2D are hyperglycemia, hyperlipidemia, insulin resistance, and impaired insulin secretion. Patients with diabetes and myocardial infarction have worse prognosis than those without T2D. Moreover, obesity and T2D are recognized risk factors in developing severe form of COVID-19 with higher mortality rate. The current lines of drug therapy are insufficient to control T2D and its serious cardiovascular complications. Phosphodiesterase 5 (PDE5) is a cGMP specific enzyme, which is the target of erectile dysfunction drugs including sildenafil, vardenafil, and tadalafil. Cardioprotective effects of PDE5 inhibitors against ischemia/reperfusion (I/R) injury were reported in normal and diabetic animals. Hydroxychloroquine (HCQ) is a widely used antimalarial and anti-inflammatory drug and its hyperglycemia-controlling effect in diabetic patients is also under investigation. This review provides our perspective of a potential use of combination therapy of PDE5 inhibitor with HCQ to reduce cardiovascular risk factors and myocardial I/R injury in T2D. We previously observed that diabetic mice treated with tadalafil and HCQ had significantly reduced fasting blood glucose and lipid levels, increased plasma insulin and insulin-like growth factor-1 levels, and improved insulin sensitivity, along with smaller myocardial infarct size following I/R. The combination treatment activated Akt/mTOR cellular survival pathway, which was likely responsible for the salutary effects. Therefore, pretreatment with PDE5 inhibitor and HCQ may be a potentially useful therapy not only for controlling T2D but also reducing the rate and severity of COVID-19 infection in the vulnerable population of diabetics.
Collapse
Affiliation(s)
- Rakesh C Kukreja
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA.
| | - Rui Wang
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA
| | - Saisudha Koka
- Department of Microbiology, Immunology and Pharmacology, Arkansas College of Osteopathic Medicine, Fort Smith, AR, 72916-6024, USA
| | - Anindita Das
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA
| | - Arun Samidurai
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA
| | - Lei Xi
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, 1101 East Marshall Street, Room 7-020D, Box 980204, Richmond, VA, 23298-0204, USA.
| |
Collapse
|
40
|
Zhang J, Li Y, Chen Y, Yu X, Wang S, Sun H, Zheng X, Zhang L, Wang Y, Zhu D. Circ-calm4 regulates hypoxia-induced pulmonary artery smooth muscle autophagy by binding Purb. J Mol Cell Cardiol 2023; 176:41-54. [PMID: 36716953 DOI: 10.1016/j.yjmcc.2023.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/28/2023]
Abstract
Pulmonary hypertension (PH) is a serious and fatal disease characterized by pulmonary vasoconstriction and pulmonary vascular remodeling. The excessive autophagy of pulmonary artery smooth muscle cells (PASMCs) is one of the important factors of pulmonary vascular remodeling. A number of studies have shown that circular RNA (circRNA) can participate in the onset of PH. Our previous studies have shown that circRNA calmodulin 4 (circ-calm4) is involved in the progression of hypoxic PH. However, the role of circ-calm4 on regulation of hypoxic PH autophagy has not been reported. In this study, we demonstrated for the first time that hypoxia-mediated upregulated circ-calm4 expression has a key regulatory effect on autophagy in hypoxia-induced PASMCs and hypoxic PH mouse models. Knockdown of circ-calm4 both in vivo and in vitro can inhibit the autophagy in PASMCs induced by hypoxia. We also performed bioinformatics predictions and conducted experiments to verify that circ-calm4 bound to the purine-rich binding protein (Purb) to promote its expression in the nucleus, thereby initiating the transcription of autophagy-related protein Beclin1. Interestingly, we found that Beclin1 transcription initiated by Purb was accompanied by a modification of Beclin1 super-enhancer to improve transcription activity and efficiency. Overall, our results confirm that the circ-calm4/Purb/Beclin1 signal axis is involved in the occurrence of hypoxia-induced PASMCs autophagy, and the novel regulatory mechanisms and signals transduction pathways in PASMC autophagy induced by hypoxia.
Collapse
Affiliation(s)
- Junting Zhang
- College of Pharmacy, Harbin Medical University, PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Yiying Li
- College of Pharmacy, Harbin Medical University, PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Yujie Chen
- College of Pharmacy, Harbin Medical University, PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Xiufeng Yu
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Shanshan Wang
- College of Pharmacy, Harbin Medical University, PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Hanliang Sun
- College of Pharmacy, Harbin Medical University, PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Xiaodong Zheng
- Department of Pharmacology, Harbin Medical University (Daqing), PR China
| | - Lixin Zhang
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China; Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Yifan Wang
- Central Laboratory of Harbin Medical University (Daqing), PR China
| | - Daling Zhu
- College of Pharmacy, Harbin Medical University, PR China; College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China; Central Laboratory of Harbin Medical University (Daqing), PR China; State Province Key Laboratories of Biomedicine-Pharmaceutics of China, PR China; Key Laboratory of Cardiovascular Medicine Research, Ministry of Education, Harbin Medical University, PR China..
| |
Collapse
|
41
|
Piper B, Bogamuwa S, Hossain T, Farkas D, Rosas L, Green A, Newcomb G, Sun N, Horowitz JC, Bhagwani AR, Yang H, Kudryashova TV, Rojas M, Mora AL, Yan P, Mallampalli RK, Goncharova EA, Eckmann DM, Farkas L. RAB7 deficiency impairs pulmonary artery endothelial function and promotes pulmonary hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.526842. [PMID: 36778418 PMCID: PMC9915659 DOI: 10.1101/2023.02.03.526842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a devastating and progressive disease with limited treatment options. Endothelial dysfunction plays a central role in development and progression of PAH, yet the underlying mechanisms are incompletely understood. The endosome-lysosome system is important to maintain cellular health and the small GTPase RAB7 regulates many functions of this system. Here, we explored the role of RAB7 in endothelial cell (EC) function and lung vascular homeostasis. We found reduced expression of RAB7 in ECs from PAH patients. Endothelial haploinsufficiency of RAB7 caused spontaneous PH in mice. Silencing of RAB7 in ECs induced broad changes in gene expression revealed via RNA sequencing and RAB7 silenced ECs showed impaired angiogenesis, expansion of a senescent cell fraction, combined with impaired endolysosomal trafficking and degradation, which suggests inhibition of autophagy at the pre-degradation level. Further, mitochondrial membrane potential and oxidative phosphorylation were decreased, and glycolysis was enhanced. Treatment with the RAB7 activator ML-098 reduced established PH in chronic hypoxia/SU5416 rats. In conclusion, we demonstrate here for the first time the fundamental impairment of EC function by loss of RAB7 that leads to PH and show RAB7 activation as a potential therapeutic strategy in a preclinical model of PH.
Collapse
|
42
|
Aparicio IM, Rojo-Domínguez P, Castillejo-Rufo A, Peña FJ, Tapia JA. The Autophagy Marker LC3 Is Processed during the Sperm Capacitation and the Acrosome Reaction and Translocates to the Acrosome Where It Colocalizes with the Acrosomal Membranes in Horse Spermatozoa. Int J Mol Sci 2023; 24:ijms24020937. [PMID: 36674454 PMCID: PMC9862423 DOI: 10.3390/ijms24020937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/25/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Despite its importance in somatic cells and during spermatogenesis, little is known about the role that autophagy may play in ejaculated spermatozoa. Our aim was to investigate whether the molecular components of autophagy, such as microtubule-associated protein 1 light chain 3 (LC3), are activated in stallion spermatozoa during the capacitation and acrosome reaction and if this activation could modulate these biological processes. To analyze the autophagy turnover, LC3I and LC3II proteins were assessed by western blotting, and the ratio between both proteins (LC3II/LC3I) was calculated. In somatic cells, this ratio indicates that autophagy has been activated and similar LC3 processing has been described in mammalian spermatozoa. The subcellular localization of autophagy-related proteins was assessed by immunofluorescence with specific antibodies that recognized Atg16, Beclin-1, and LC3. The colocalization of acrosomal membranes (PNA) and LC3 was studied by confocal microcopy, and the acrosome reacted cells were quantified by flow cytometry. The incubation of stallion sperm in capacitating conditions (BWW; 3 h) significantly increased LC3 processing. This increment was three to four times higher after the induction of the acrosome reaction in these cells. LC3 was mainly expressed in the head in mature ejaculated sperm showing a clear redistribution from the post-acrosomal region to the acrosome upon the incubation of sperm in capacitating conditions (BWW, 3 h). After the induction of the acrosome reaction, LC3 colocalized with the acrosome or the apical plasmalemma membranes in the head of the stallion spermatozoa. The inhibition or activation of autophagy-related pathways in the presence of autophagy activators (STF-62247) or inhibitors (E-64d, chloroquine) significantly increased LC3 processing and increased the percent of acrosome reacted cells, whereas 3-methyladenine almost completely inhibited LC3 processing and the acrosome reaction. In conclusion, we found that sperm capacitation and acrosome reaction could be regulated by autophagy components in sperm cells ex vivo by processes that might be independent of the intraluminal pH of the acrosome and dependent of LC3 lipidation. It can be speculated that, in stallion sperm, a form of noncanonical autophagy utilizes some components of autophagy machinery to facilitate the acrosome reaction.
Collapse
Affiliation(s)
- Ines M. Aparicio
- Department of Physiology, Institute of Molecular Pathology Biomarkers (BICOMCEL), University of Extremadura, 10003 Cáceres, Spain
| | - Patricia Rojo-Domínguez
- Laboratory of Spermatology, Veterinary Teaching Hospital, University of Extremadura, 10003 Cáceres, Spain
| | - Alba Castillejo-Rufo
- Department of Physiology, Institute of Molecular Pathology Biomarkers (BICOMCEL), University of Extremadura, 10003 Cáceres, Spain
| | - Fernando J. Peña
- Laboratory of Spermatology, Veterinary Teaching Hospital, University of Extremadura, 10003 Cáceres, Spain
| | - Jose A. Tapia
- Department of Physiology, Institute of Molecular Pathology Biomarkers (BICOMCEL), University of Extremadura, 10003 Cáceres, Spain
- Correspondence:
| |
Collapse
|
43
|
Bao C, Liang S, Han Y, Yang Z, Liu S, Sun Y, Zheng S, Li Y, Wang T, Gu Y, Wu K, Black SM, Wang J, Nawrocki ST, Carew JS, Yuan JXJ, Tang H. The Novel Lysosomal Autophagy Inhibitor (ROC-325) Ameliorates Experimental Pulmonary Hypertension. Hypertension 2023; 80:70-83. [PMID: 36345832 DOI: 10.1161/hypertensionaha.122.19397] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Autophagy plays an important role in the pathogenesis of pulmonary hypertension (PH). ROC-325 is a novel small molecule lysosomal autophagy inhibitor that has more potent anticancer activity than the antimalarial drug hydroxychloroquine, the latter has been prevalently used to inhibit autophagy. Here, we sought to determine the therapeutic benefit and mechanism of action of ROC-325 in experimental PH models. METHODS AND RESULTS Hemodynamics, echocardiography, and histology measurement showed that ROC-325 treatment prevented the development of PH, right ventricular hypertrophy, fibrosis, dysfunction, and vascular remodeling after monocrotaline and Sugen5416/hypoxia administration. ROC-325 attenuated high K+ or alveolar hypoxia-induced pulmonary vasoconstriction and enhanced endothelial-dependent relaxation in isolated pulmonary artery rings. ROC-325 treatment inhibited autophagy and enhanced endothelial nitric oxide synthase activity in lung tissues of monocrotaline-PH rats. In cultured human and rat pulmonary arterial smooth muscle cell and pulmonary arterial endothelial cell under hypoxia exposure, ROC-325 increased LC3B (light chain 3 beta) and p62 accumulation, endothelial cell nitric oxide production via phosphorylation of endothelial nitric oxide synthase (Ser1177) and dephosphorylation of endothelial nitric oxide synthase (Thr495) as well as decreased HIF (hypoxia-inducible factor)-1α and HIF-2α stabilization. CONCLUSIONS These data indicate that ROC-325 is a promising novel agent for the treatment of PH that inhibits autophagy, downregulates HIF levels, and increases nitric oxide production.
Collapse
Affiliation(s)
- Changlei Bao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.).,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China (C.B., S.L., Y.S., S.Z.)
| | - Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.)
| | - Ying Han
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, China (Y.H.)
| | - Zi Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.)
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.).,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China (C.B., S.L., Y.S., S.Z.)
| | - Yanan Sun
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.).,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China (C.B., S.L., Y.S., S.Z.)
| | - Shichuang Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.).,College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China (C.B., S.L., Y.S., S.Z.)
| | - Yuzhu Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.)
| | - Ting Wang
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Miami, FL (T.W., S.M.B.).,Center for Translational Science and Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Port St. Lucie, FL (T.W., S.M.B.)
| | - Yali Gu
- Banner University of Arizona Medical Center, Tucson, AZ (Y.G.)
| | - Kang Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.)
| | - Stephen M Black
- Department of Cellular Biology and Pharmacology, Herbert Wertheim College of Medicine, Miami, FL (T.W., S.M.B.).,Center for Translational Science and Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Port St. Lucie, FL (T.W., S.M.B.)
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.)
| | | | - Jennifer S Carew
- University of Arizona Cancer Center, Tucson, AZ (S.T.N., J.S.C.)
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, La Jolla, CA (J.X.-J.)
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, China (C.B., S.L., Z.Y., S.L., Y.S., S.Z., Y.L., K.W., J.W., H.T.)
| |
Collapse
|
44
|
Chen Y, Bao M, Liu JT, Bao H, Zhang SM, Lou Y, Qi YX. Defective autophagy triggered by arterial cyclic stretch promotes neointimal hyperplasia in vein grafts via the p62/nrf2/slc7a11 signaling pathway. J Mol Cell Cardiol 2022; 173:101-114. [PMID: 36308866 DOI: 10.1016/j.yjmcc.2022.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/21/2022]
Abstract
Autophagy is an adaptation mechanism to keep cellular homeostasis, and its deregulation is implicated in various cardiovascular diseases. After vein grafting, hemodynamic factors play crucial roles in neointimal hyperplasia, but the mechanisms are poorly understood. Here, we investigated the impacts of arterial cyclic stretch on autophagy of venous smooth muscle cells (SMCs) and its role in neointima formation after vein grafting. Rat jugular vein graft were generated via the 'cuff' technique. Autophagic flux in venous SMCs is impaired in 3-day, 1-week and 2-week grafted veins. 10%-1.25 Hz cyclic stretch (arterial stretch) loaded with FX5000 stretch system on venous SMCs blocks cellular autophagic flux in vitro and shows no significant impact on activity of mTORC1 and AMPK. Microtubule depolymerization but not lysosome dysfunction nor autophagosome/amphisome-lysosomal membrane fusion blockade is involved in the impairment of autophagic flux. Microtubule stabilization, induced by paclitaxel treatment and external stents intervention respectively, restores venous SMC autophagy and ameliorates neointimal hyperplasia in vivo. Moreover, autophagy impairment causes accumulation of the cargo receptor p62, which sequesters keap1 to p62 aggregates and results in the stabilization and nuclear translocation of nrf2 to modulate its target antioxidative gene SLC7A11. p62 silencing abrogates the increases of nrf2 and slc7a11 protein expression, glutathione level and venous SMC proliferation triggered by arterial cyclic stretch in vitro, and further hinders nrf2 nuclear translocation, reduces neointimal thickness after vein grafting in vivo. p62 (T349A) mutation also inhibited venous SMC proliferation and alleviated neointimal formation in vivo. These findings suggest that stabilization of microtubules to rescue autophagic flux or direct silencing of p62 are potential therapeutic strategies for neointimal hyperplasia.
Collapse
Affiliation(s)
- Yi Chen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Min Bao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ji-Ting Liu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Han Bao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shou-Min Zhang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yue Lou
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
45
|
Choi M, Hwang JR, Sung JH, Byun N, Seok YS, Cho GJ, Choi SJ, Kim JS, Oh SY, Roh CR. Hydroxychloroquine reduces hypertension and soluble fms-like kinase-1 in a Nω-nitro-l-arginine methyl ester-induced preeclampsia rat model. J Hypertens 2022; 40:2459-2468. [PMID: 36321404 DOI: 10.1097/hjh.0000000000003279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
OBJECTIVE Hydroxychloroquine, a drug used for malaria and autoimmune diseases reportedly has beneficial effects against preeclampsia in pregnant women with lupus. However, its mechanism against preeclampsia remains unclear. We investigated the effect of hydroxychloroquine on an Nω-nitro-l-arginine methyl ester-induced preeclampsia rat model. METHODS Pregnant Sprague-Dawley rats were divided into four groups based on treatment (administered on gestational days 7-18): control, Nω-nitro-l-arginine methyl ester, hydroxychloroquine, and Nω-nitro-l-arginine methyl ester plus hydroxychloroquine. All animals were sacrificed on gestational day 19. We assayed tube formation and determined reactive oxygen species levels using human umbilical vein endothelial cells. RESULTS Results showed that hydroxychloroquine significantly lowered mean systolic blood pressure (P < 0.05) in Nω-nitro-l-arginine methyl ester-treated rats. Hydroxychloroquine did not affect their fetal and placental weights. Hydroxychloroquine mitigated Nω-nitro-l-arginine methyl ester-associated changes in proteinuria (P < 0.05). It normalized plasma soluble fms-like kinase-1 (P < 0.05) and endothelin-1 (P < 0.01) levels. In the tube formation assay, hydroxychloroquine increased the total meshes area (P < 0.05) and mitigated Nω-nitro-l-arginine methyl ester-induced reactive oxygen species formation (P < 0.05) in human umbilical vein endothelial cells. CONCLUSION We conclude that hydroxychloroquine alleviated hypertension, proteinuria, and normalized soluble fms-like kinase-1 and endothelin-1 levels in our preeclampsia model and that these changes may involve the restoration of endothelial dysfunction; thus, hydroxychloroquine could potentially be used for preventing preeclampsia, even in the absence of lupus.
Collapse
Affiliation(s)
- Minji Choi
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University
- Samsung Biomedical Research Institute, Samsung Medical Center
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Jae Ryoung Hwang
- Sungkyunkwan University School of Medicine, Samsung Biomedical Research Institute, Samsung Medical Center
| | - Ji-Hee Sung
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Nagyeong Byun
- Samsung Biomedical Research Institute, Samsung Medical Center
| | - Ye Seon Seok
- Department of Obstetrics and Gynecology, Korea University College of Medicine
| | - Geum Joon Cho
- Department of Obstetrics and Gynecology, Korea University College of Medicine
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Jung-Sun Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine
| | - Cheong-Rae Roh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine
| |
Collapse
|
46
|
Devendran A, Kar S, Bailey R, Trivieri MG. The Role of Bone Morphogenetic Protein Receptor Type 2 ( BMPR2) and the Prospects of Utilizing Induced Pluripotent Stem Cells (iPSCs) in Pulmonary Arterial Hypertension Disease Modeling. Cells 2022; 11:3823. [PMID: 36497082 PMCID: PMC9741276 DOI: 10.3390/cells11233823] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by increased pulmonary vascular resistance (PVR), causing right ventricular hypertrophy and ultimately death from right heart failure. Heterozygous mutations in the bone morphogenetic protein receptor type 2 (BMPR2) are linked to approximately 80% of hereditary, and 20% of idiopathic PAH cases, respectively. While patients carrying a BMPR2 gene mutation are more prone to develop PAH than non-carriers, only 20% will develop the disease, whereas the majority will remain asymptomatic. PAH is characterized by extreme vascular remodeling that causes pulmonary arterial endothelial cell (PAEC) dysfunction, impaired apoptosis, and uncontrolled proliferation of the pulmonary arterial smooth muscle cells (PASMCs). To date, progress in understanding the pathophysiology of PAH has been hampered by limited access to human tissue samples and inadequacy of animal models to accurately mimic the pathogenesis of human disease. Along with the advent of induced pluripotent stem cell (iPSC) technology, there has been an increasing interest in using this tool to develop patient-specific cellular models that precisely replicate the pathogenesis of PAH. In this review, we summarize the currently available approaches in iPSC-based PAH disease modeling and explore how this technology could be harnessed for drug discovery and to widen our understanding of the pathophysiology of PAH.
Collapse
Affiliation(s)
- Anichavezhi Devendran
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumanta Kar
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rasheed Bailey
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maria Giovanna Trivieri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Cardiology Unit, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
47
|
Ainscough AJ, Smith TJ, Haensel M, Rhodes CJ, Fellows A, Whitwell HJ, Vasilaki E, Gray K, Freeman A, Howard LS, Wharton J, Dunmore B, Upton PD, Wilkins MR, Edel JB, Wojciak-Stothard B. An organ-on-chip model of pulmonary arterial hypertension identifies a BMPR2-SOX17-prostacyclin signalling axis. Commun Biol 2022; 5:1192. [PMID: 36344664 PMCID: PMC9640600 DOI: 10.1038/s42003-022-04169-z] [Citation(s) in RCA: 7] [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: 05/04/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an unmet clinical need. The lack of models of human disease is a key obstacle to drug development. We present a biomimetic model of pulmonary arterial endothelial-smooth muscle cell interactions in PAH, combining natural and induced bone morphogenetic protein receptor 2 (BMPR2) dysfunction with hypoxia to induce smooth muscle activation and proliferation, which is responsive to drug treatment. BMPR2- and oxygenation-specific changes in endothelial and smooth muscle gene expression, consistent with observations made in genomic and biochemical studies of PAH, enable insights into underlying disease pathways and mechanisms of drug response. The model captures key changes in the pulmonary endothelial phenotype that are essential for the induction of SMC remodelling, including a BMPR2-SOX17-prostacyclin signalling axis and offers an easily accessible approach for researchers to study pulmonary vascular remodelling and advance drug development in PAH.
Collapse
Affiliation(s)
- Alexander J Ainscough
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Chemistry, Imperial College London, London, UK
| | - Timothy J Smith
- Department of Chemistry, Imperial College London, London, UK
| | - Maike Haensel
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Adam Fellows
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Harry J Whitwell
- National Phenome Centre and Imperial Clinical Phenotyping Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Section of Bioanalytical Chemistry, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Eleni Vasilaki
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Kelly Gray
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Adrian Freeman
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Benjamin Dunmore
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Paul D Upton
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Royal Papworth Hospitals, Cambridge, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Joshua B Edel
- Department of Chemistry, Imperial College London, London, UK
| | | |
Collapse
|
48
|
Chen S, Wei X, Zhang X, Yao M, Qiu Z, Chen L, Zhang L. Supplementation with Tex261 provides a possible preventive treatment for hypoxic pulmonary artery hypertension. Front Pharmacol 2022; 13:1028058. [PMID: 36408272 PMCID: PMC9669906 DOI: 10.3389/fphar.2022.1028058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/19/2022] [Indexed: 10/28/2023] Open
Abstract
Objectives: Pulmonary artery hypertension (PAH) is a serious disease for which there is no effective treatment. Its pathogenesis is complex and has not yet been clarified. Tex261 is a protein-coding gene whose functional enrichment nodes include the transporter activity of COP II. However, the role of Tex261 in PAH remains unknown. Methods: Sugen5416/Hypoxic PAH models were established, and pulmonary arteries (PAs) were isolated for proteomic sequencing. The binding sites between Hif-1α and Tex261 were verified by dual-luciferase reporter gene assay. Cell proliferation was detected by MTS and EdU assays. For determination of the preventive and therapeutic effects of Tex261, intratracheal instillation of adeno-associated virus (AVV6) with Tex261 vectors was performed. Results: Tex261 was screened according to the proteomic sequencing data. Hif-1α inhibited Tex261 promoter activity under hypoxia. Decreased Tex261 expression promoted PASMC proliferation. Tex261 regulated Sec23 via the Ndrg1-mediated Akt pathway. Tex261 overexpression improved the pressure and vessel remodeling of PAs induced by Sugen5416/hypoxia. Conclusion: Hypoxia suppressed Tex261 expression through Hif-1α activation. The decreased Tex261 could promote Ndrg1 and depress Akt activity and then inhibit Sec23 activity, which leads to cell proliferation and vessel remodeling. Elevated Tex261 has some preventive and therapeutic effects on rats with PAH.
Collapse
Affiliation(s)
- Shaokun Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Pathophysiology, The School of Basic Medical Sciences, The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fuzhou, China
| | - Xiaozhen Wei
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Pathophysiology, The School of Basic Medical Sciences, The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fuzhou, China
| | - Xu Zhang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Pathophysiology, The School of Basic Medical Sciences, The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fuzhou, China
| | - Mengge Yao
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Pathophysiology, The School of Basic Medical Sciences, The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fuzhou, China
| | - Zhihuang Qiu
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Liangwan Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Li Zhang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Pathophysiology, The School of Basic Medical Sciences, The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Fuzhou, China
| |
Collapse
|
49
|
Wu Q, Zhou X, Wang Y, Hu Y. LncRNA GAS5 promotes spermidine‑induced autophagy through the miRNA‑31‑5p/NAT8L axis in pulmonary artery endothelial cells of patients with CTEPH. Mol Med Rep 2022; 26:297. [PMID: 35920180 PMCID: PMC9434988 DOI: 10.3892/mmr.2022.12813] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/23/2021] [Indexed: 11/17/2022] Open
Abstract
Chronic thromboembolic pulmonary hypertension (CTEPH) is a leading cause of pulmonary hypertension. The present study investigated the mechanisms of long non-coding RNA growth arrest-specific transcript 5 (GAS5) on spermidine (SP)-induced autophagy. Pulmonary artery endothelial cells (PAECs) were collected from patients with CTEPH and the rat model. Immunofluorescence, Western blots, reverse transcription-quantitative polymerase chain reaction, bioinformatics, rapid amplification of cDNA ends assays, luciferase reporter assays, RNA-binding protein immunoprecipitation assays, GFP-LC3 adenoviruses, tfLC3 assays and transmission electron microscopy were performed. The results revealed that SP-induced autophagy increased GAS5 in PAECs. The upregulation of GAS5 enhanced and the downregulation of GAS5 reversed the roles of SP in PAECs. Furthermore, GAS5 promoted SP-induced autophagy in PAECs by targeting miRNA-31-5p. The miRNA-31-5p mimic suppressed and the inhibitor promoted SP-induced autophagy. Furthermore, N-Acetyltransferase 8 Like (NAT8L) was a target gene of miRNA-31-5p and knockdown of NAT8L inhibited the autophagic levels of PAECs. In vivo, SP treatment decreased miRNA-31-5p and increased NAT8L levels, which was reversed by the knockdown of GAS5. The downregulation of GAS5 abolished the stimulatory role of SP in PAECs of CTEPH rats. In conclusion, GAS5 promoted SP-induced autophagy through miRNA-31-5p/NAT8L signaling pathways in vitro and in vivo and GAS5 may be a promising molecular marker for therapies of CTEPH.
Collapse
Affiliation(s)
- Qinghua Wu
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Xiaohui Zhou
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yan Wang
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Yamin Hu
- Department of Cardiovascular Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| |
Collapse
|
50
|
Zhai C, Zhang N, Wang J, Cao M, Luan J, Liu H, Zhang Q, Zhu Y, Xue Y, Li S. Activation of Autophagy Induces Monocrotaline-Induced Pulmonary Arterial Hypertension by FOXM1-Mediated FAK Phosphorylation. Lung 2022; 200:619-631. [PMID: 36107242 DOI: 10.1007/s00408-022-00569-4] [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: 04/12/2022] [Accepted: 09/01/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE It has been shown that activation of autophagy promotes the development of pulmonary arterial hypertension (PAH). Meanwhile, forkhead box M1 (FOXM1) has been found to induce autophagy in several types of cancer. However, it is still unclear whether FOXM1 mediates autophagy activation in PAH, and detailed mechanisms responsible for these processes are indefinite. METHOD PAH was induced by a single intraperitoneal injection of monocrotaline (MCT) to rats. The right ventricle systolic pressure (RVSP), right ventricular hypertrophy index (RVHI), percentage of medial wall thickness (%MT), α-smooth muscle actin (α-SMA) staining, and Ki67 staining were performed to evaluate the development of PAH. The protein levels of FOXM1, phospho-focal adhesion kinase (p-FAK), FAK, and LC3B were determined by immunoblotting or immunohistochemistry. RESULTS FOXM1 protein level and FAK activity were significantly increased in MCT-induced PAH rats, this was accompanied with the activation of autophagy. Pharmacological inhibition of FOXM1 or FAK suppressed MCT-induced autophagy activation, decreased RVSP, RVHI and %MT in MCT-induced PAH rats, and inhibited the proliferation of pulmonary arterial smooth muscle cells and pulmonary vessel muscularization in MCT-induced PAH rats. CONCLUSION FOXM1 promotes the development of PAH by inducing FAK phosphorylation and subsequent activation of autophagy in MCT-treated rats.
Collapse
Affiliation(s)
- Cui Zhai
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Nana Zhang
- Center for Regenerative and Reconstructive Medicine, Med-X Institute of the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Meng Cao
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Jing Luan
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Huan Liu
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China
| | - Yanting Zhu
- Center of Nephropathy and Hemodialysis, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, People's Republic of China
| | - Yuxin Xue
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, 710021, Shaanxi, People's Republic of China
| | - Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No.277, West Yanta Road, Xi'an, 710061, Shaanxi, People's Republic of China.
| |
Collapse
|