1
|
Shen YH, Ding D, Lian TY, Qiu BC, Yan Y, Wang PW, Zhang WH, Jing ZC. Panorama of artery endothelial cell dysfunction in pulmonary arterial hypertension. J Mol Cell Cardiol 2024; 197:61-77. [PMID: 39437884 DOI: 10.1016/j.yjmcc.2024.10.004] [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: 05/11/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal lung disease characterized by progressive pulmonary vascular remodeling. The initial cause of pulmonary vascular remodeling is the dysfunction of pulmonary arterial endothelial cells (PAECs), manifested by changes in the categorization of cell subtypes, endothelial programmed cell death, such as apoptosis, necroptosis, pyroptosis, ferroptosis, et al., overproliferation, senescence, metabolic reprogramming, endothelial-to-mesenchymal transition, mechanosensitivity, and regulation ability of peripheral cells. Therefore, it is essential to explore the mechanism of endothelial dysfunction in the context of PAH. This review aims to provide a comprehensive understanding of the molecular mechanisms underlying endothelial dysfunction in PAH. We highlight the developmental process of PAECs and changes in PAH and summarise the latest classification of endothelial dysfunction. Our review could offer valuable insights into potential novel EC-specific targets for preventing and treating PAH.
Collapse
Affiliation(s)
- Ying-Huizi Shen
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Dong Ding
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian-Yu Lian
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bao-Chen Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Yan
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei-Wen Wang
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei-Hua Zhang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Meng ZY, Lu CH, Li J, Liao J, Wen H, Li Y, Huang F, Zeng ZY. Identification and experimental verification of senescence-related gene signatures and molecular subtypes in idiopathic pulmonary arterial hypertension. Sci Rep 2024; 14:22157. [PMID: 39333589 PMCID: PMC11437103 DOI: 10.1038/s41598-024-72979-8] [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: 06/11/2024] [Accepted: 09/12/2024] [Indexed: 09/29/2024] Open
Abstract
Evidences illustrate that cell senescence contributes to the development of pulmonary arterial hypertension. However, the molecular mechanisms remain unclear. Since there may be different senescence subtypes between PAH patients, consistent senescence-related genes (SRGs) were utilized for consistent clustering by unsupervised clustering methods. Senescence is inextricably linked to the immune system, and the immune cells in each cluster were estimated by ssGSEA. To further screen out more important SRGs, machine learning algorithms were used for identification and their diagnostic value was assessed by ROC curves. The expression of hub genes were verified in vivo and in vitro. Transcriptome analysis was used to assess the effects of silence of hub gene on different pathways. Three senescence molecular subtypes were identified by consensus clustering. Compared with cluster A and B, most immune cells and checkpoint genes were higher in cluster C. Thus, we identified senescence cluster C as the immune subtype. The ROC curves of IGF1, HOXB7, and YWHAZ were remarkable in both datasets. The expression of these genes was increased in vitro. Western blot and immunohistochemical analyses revealed that YWHAZ expression was also increased. Our transcriptome analysis showed autophagy-related genes were significantly elevated after silence of YWHAZ. Our research provided several prospective SRGs and molecular subtypes. Silence of YWHAZ may contribute to the clearance of senescent endothelial cells by activating autophagy.
Collapse
Affiliation(s)
- Zhong-Yuan Meng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Chuang-Hong Lu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jing Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Juan Liao
- Ultrasound Department, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Hong Wen
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yuan Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Feng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| | - Zhi-Yu Zeng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, No.6 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
3
|
Chu Q, Li Y, Wu J, Gao Y, Guo X, Li J, Lv H, Liu M, Tang W, Zhan P, Zhang T, Hu H, Liu H, Sun J, Wang X, Yi F. Oxysterol Sensing Through GPR183 Triggers Endothelial Senescence in Hypertension. Circ Res 2024; 135:708-721. [PMID: 39176657 DOI: 10.1161/circresaha.124.324722] [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: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Despite endothelial dysfunction being an initial step in the development of hypertension and associated cardiovascular/renal injuries, effective therapeutic strategies to prevent endothelial dysfunction are still lacking. GPR183 (G protein-coupled receptor 183), a recently identified G protein-coupled receptor for oxysterols and hydroxylated metabolites of cholesterol, has pleiotropic roles in lipid metabolism and immune responses. However, the role of GPR183 in the regulation of endothelial function remains unknown. METHODS Endothelial-specific GPR183 knockout mice were generated and used to examine the role of GPR183 in endothelial senescence by establishing 2 independent hypertension models: desoxycorticosterone acetate/salt-induced and Ang II (angiotensin II)-induced hypertensive mice. Echocardiography, transmission electron microscopy, blood pressure measurement, vasorelaxation response experiments, flow cytometry analysis, and chromatin immunoprecipitation analysis were performed in this study. RESULTS Endothelial GPR183 was significantly induced in hypertensive mice, which was further confirmed in renal biopsies from subjects with hypertensive nephropathy. Endothelial-specific deficiency of GPR183 markedly alleviated cardiovascular and renal injuries in hypertensive mice. Moreover, we found that GPR183 regulated endothelial senescence in both hypertensive mice and aged mice. Mechanistically, GPR183 disrupted circadian signaling by inhibiting PER1 (period circadian regulator 1) expression, thereby facilitating endothelial senescence and dysfunction through the cAMP (cyclic adenosine monophosphate)/PKA (protein kinase A)/CREB (cAMP-response element binding protein) signaling pathway. Importantly, pharmacological inhibition of the oxysterol-GPR183 axis by NIBR189 or clotrimazole ameliorated endothelial senescence and cardiovascular/renal injuries in hypertensive mice. CONCLUSIONS This study discovers a previously unrecognized role of GPR183 in promoting endothelial senescence. Pharmacological targeting of GPR183 may be an innovative therapeutic strategy for hypertension and its associated complications.
Collapse
Affiliation(s)
- Qingqing Chu
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Yujia Li
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital (Y.L., F.Y.), Shandong University, Jinan, China
| | - Jichao Wu
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Yanjiao Gao
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Xiangyun Guo
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Jing Li
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Hang Lv
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Min Liu
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Wei Tang
- Department of Pathogenic Biology, School of Basic Medical Sciences (W.T.), Shandong University, Jinan, China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences (P.Z.), Shandong University, Jinan, China
| | - Tao Zhang
- Department of Biostatistics, School of Public Health (T.Z.), Shandong University, Jinan, China
| | - Huili Hu
- Department of Systems Biomedicine and Research Center of Stem Cell and Regenerative Medicine, School of Basic Medical Sciences (H.H.), Shandong University, Jinan, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials (H. Liu), Shandong University, Jinan, China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences (J.S.), Shandong University, Jinan, China
| | - Xiaojie Wang
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences (Q.C., Y.L., J.W., Y.G., X.G., J.L., H. Lv, M.L., X.W., F.Y.), Shandong University, Jinan, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital (Y.L., F.Y.), Shandong University, Jinan, China
| |
Collapse
|
4
|
Guo Y, Cheng X, Huang C, Gao J, Shen W. Frataxin Loss Promotes Angiotensin II-Induced Endothelial-to-Mesenchymal Transition. J Am Heart Assoc 2024; 13:e034316. [PMID: 39023059 DOI: 10.1161/jaha.124.034316] [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: 03/26/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND The metabolic flexibility of endothelial cells is linked to their phenotypic plasticity. Frataxin is critical in determining the iron metabolism and fate of endothelial cells. This study aimed to investigate frataxin-mediated metabolic remodeling during the endothelial-to-mesenchymal transition (EndoMT). METHODS AND RESULTS Endothelial cell-specific frataxin knockout and frataxin mutation mice were subjected to angiotensin II to induce hypertension. EndoMT and cardiac fibrosis were assessed using histological and protein expression analyses. Fatty acid oxidation (FAO) in microvascular endothelial cells was measured using a Seahorse XF96 analyzer. We showed that inhibition of FAO accompanies angiotensin II-induced EndoMT. Frataxin knockout mice promote EndoMT, associated with increased cardiac fibrosis following angiotensin II infusion. Angiotensin II reduces frataxin expression, which leads to mitochondrial iron overload and subsequent carbonylation of sirtuin 3. In turn, carbonylated sirtuin 3 contributes to the acetylated frataxin at lysine 189, making it more prone to degradation. The frataxin/sirtuin 3 feedback loop reduces hydroxyl-CoA dehydrogenase α subunit-mediated FAO. Additionally, silymarin is a scavenger of free radicals, restoring angiotensin II-induced reduction of FAO activity and sirtuin 3 and frataxin expression, improving EndoMT both in vitro and in vivo. Furthermore, frataxin mutation mice showed suppressed EndoMT and improved cardiac fibrosis. CONCLUSIONS The frataxin/sirtuin 3 feedback loop has the potential to attenuate angiotensin II-induced EndoMT by improving FAO.
Collapse
Affiliation(s)
- Yuetong Guo
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xingyi Cheng
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Chenglin Huang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jing Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Weili Shen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
5
|
Safaie Qamsari E, Stewart DJ. Cellular senescence in the pathogenesis of pulmonary arterial hypertension: the good, the bad and the uncertain. Front Immunol 2024; 15:1403669. [PMID: 39156894 PMCID: PMC11329925 DOI: 10.3389/fimmu.2024.1403669] [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: 03/19/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
Senescence refers to a cellular state marked by irreversible cell cycle arrest and the secretion of pro-inflammatory and tissue-remodeling factors. The senescence associated secretory phenotype (SASP) impacts the tissue microenvironment and provides cues for the immune system to eliminate senescent cells (SCs). Cellular senescence has a dual nature; it can be beneficial during embryonic development, tissue repair, and tumor suppression, but it can also be detrimental in the context of chronic stress, persistent tissue injury, together with an impairment in SC clearance. Recently, the accumulation of SCs has been implicated in the pathogenesis of pulmonary arterial hypertension (PAH), a progressive condition affecting the pre-capillary pulmonary arterial bed. PAH is characterized by endothelial cell (EC) injury, inflammation, and proliferative arterial remodeling, which leads to right heart failure and premature mortality. While vasodilator therapies can improve symptoms, there are currently no approved treatments capable of reversing the obliterative arterial remodeling. Ongoing endothelial injury and dysfunction is central to the development of PAH, perpetuated by hemodynamic perturbation leading to pathological intimal shear stress. The precise role of senescent ECs in PAH remains unclear. Cellular senescence may facilitate endothelial repair, particularly in the early stages of disease. However, in more advanced disease the accumulation of senescent ECs may promote vascular inflammation and occlusive arterial remodeling. In this review, we will examine the evidence that supports a role of endothelial cell senescence to the pathogenesis of PAH. Furthermore, we will compare and discuss the apparent contradictory outcomes with the use of interventions targeting cellular senescence in the context of experimental models of pulmonary hypertension. Finally, we will attempt to propose a framework for the understanding of the complex interplay between EC injury, senescence, inflammation and arterial remodeling, which can guide further research in this area and the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Elmira Safaie Qamsari
- Sinclair Centre for Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Duncan J. Stewart
- Sinclair Centre for Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Division of Cardiology, University of Ottawa Heart Institute, Ottawa, ON, Canada
| |
Collapse
|
6
|
Li X, Tan J, Wan J, Cheng B, Wang YH, Dai A. Cell Death in Pulmonary Arterial Hypertension. Int J Med Sci 2024; 21:1840-1851. [PMID: 39113898 PMCID: PMC11302558 DOI: 10.7150/ijms.93902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/22/2024] [Indexed: 08/10/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe pulmonary vascular disease characterized by increased pulmonary vascular resistance because of vascular remodeling and vasoconstriction. Subsequently, PAH leads to right ventricular hypertrophy and heart failure. Cell death mechanisms play a significant role in development and tissue homeostasis, and regulate the balance between cell proliferation and differentiation. Several basic and clinical studies have demonstrated that multiple mechanisms of cell death, including pyroptosis, apoptosis, autophagy, ferroptosis, anoikis, parthanatos, and senescence, are closely linked with the pathogenesis of PAH. This review summarizes different cell death mechanisms involved in the death of pulmonary artery smooth muscle cells (PASMCs) and pulmonary artery endothelial cells (PAECs), the primary target cells in PAH. This review summarizes the role of these cell death mechanisms, associated signaling pathways, unique effector molecules, and various pro-survival or reprogramming mechanisms. The aim of this review is to summarize the currently known molecular mechanisms underlying PAH. Further investigations of the cell death mechanisms may unravel new avenues for the prevention and treatment of PAH.
Collapse
Affiliation(s)
- Xia Li
- Hunan Academy of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - JunLan Tan
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - JiaJing Wan
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - BeiBei Cheng
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| | - Yu-Hong Wang
- Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Aiguo Dai
- Hunan Academy of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Department of Respiratory Diseases, Medical School, Hunan University of Chinese Medicine, Changsha 410208, Hunan, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha 410208, Hunan, People's Republic of China
| |
Collapse
|
7
|
Lei W, Chen MH, Huang ZF, Chen XY, Wang JX, Zheng J, Zhu YZ, Lan XZ, He Y. Salidroside protects pulmonary artery endothelial cells against hypoxia-induced apoptosis via the AhR/NF-κB and Nrf2/HO-1 pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155376. [PMID: 38503152 DOI: 10.1016/j.phymed.2024.155376] [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: 08/10/2023] [Revised: 12/18/2023] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND The apoptosis of pulmonary artery endothelial cells (PAECs) is an important factor contributing to the development of pulmonary hypertension (PH), a serious cardio-pulmonary vascular disorder. Salidroside (SAL) is a bioactive compound derived from an herb Rhodiola, but the potential protective effects of SAL on PAECs and the underlying mechanisms remain elusive. PURPOSE The objective of this study was to determine the role of SAL in the hypoxia-induced apoptosis of PAECs and to dissect the underlying mechanisms. STUDY DESIGN Male Sprague-Dawley (SD) rats were subjected to hypoxia (10% O2) for 4 weeks to establish a model of PH. Rats were intraperitoneally injected daily with SAL (2, 8, and 32 mg/kg/d) or vehicle. To define the molecular mechanisms of SAL in PAECs, an in vitro model of hypoxic cell injury was also generated by exposed PAECs to 1% O2 for 48 h. METHODS Various techniques including hematoxylin and eosin (HE) staining, immunofluorescence, flow cytometry, CCK-8, Western blot, qPCR, molecular docking, and surface plasmon resonance (SPR) were used to determine the role of SAL in rats and in PAECs in vitro. RESULTS Hypoxia stimulation increases AhR nuclear translocation and activates the NF-κB signaling pathway, as evidenced by upregulated expression of CYP1A1, CYP1B1, IL-1β, and IL-6, resulting in oxidative stress and inflammatory response and ultimately apoptosis of PAECs. SAL inhibited the activation of AhR and NF-κB, while promoted the nuclear translocation of Nrf2 and increased the expression of its downstream antioxidant proteins HO-1 and NQO1 in PAECs, ameliorating the hypoxia-induced oxidative stress in PAECs. Furthermore, SAL lowered right ventricular systolic pressure, and decreased pulmonary vascular remodeling and right ventricular hypertrophy in hypoxia-exposed rats. CONCLUSIONS SAL may attenuate the apoptosis of PAECs by suppressing NF-κB and activating Nrf2/HO-1 pathways, thereby delaying the progressive pathology of PH.
Collapse
Affiliation(s)
- Wei Lei
- TAAHC-GDMU Biomedical and Health Joint R&D Center, The Provincial and Ministerial Co-founded Collaborative Innovation Center for R&D in Tibet Characteristic Agriculture and Animal Husbandry Resources, The Center for Xizang Chinese (Tibetan) Medicine Resource, Joint Laboratory for Tibetan Materia Medica Resource Scientific Protection and Utilization, Tibetan Medical Research Center of Tibet, Tibet Agriculture and Animal Husbandry University, Nyingchi, Tibet, PR China; Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, GDMU-TAAHC Biomedical and Health Joint R&D Center, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Mei-Hong Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, GDMU-TAAHC Biomedical and Health Joint R&D Center, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China; Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Zu-Feng Huang
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Xiao-Ying Chen
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Jin-Xia Wang
- TAAHC-GDMU Biomedical and Health Joint R&D Center, The Provincial and Ministerial Co-founded Collaborative Innovation Center for R&D in Tibet Characteristic Agriculture and Animal Husbandry Resources, The Center for Xizang Chinese (Tibetan) Medicine Resource, Joint Laboratory for Tibetan Materia Medica Resource Scientific Protection and Utilization, Tibetan Medical Research Center of Tibet, Tibet Agriculture and Animal Husbandry University, Nyingchi, Tibet, PR China; Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Yi-Zhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, PR China
| | - Xiao-Zhong Lan
- TAAHC-GDMU Biomedical and Health Joint R&D Center, The Provincial and Ministerial Co-founded Collaborative Innovation Center for R&D in Tibet Characteristic Agriculture and Animal Husbandry Resources, The Center for Xizang Chinese (Tibetan) Medicine Resource, Joint Laboratory for Tibetan Materia Medica Resource Scientific Protection and Utilization, Tibetan Medical Research Center of Tibet, Tibet Agriculture and Animal Husbandry University, Nyingchi, Tibet, PR China.
| | - Yuan He
- TAAHC-GDMU Biomedical and Health Joint R&D Center, The Provincial and Ministerial Co-founded Collaborative Innovation Center for R&D in Tibet Characteristic Agriculture and Animal Husbandry Resources, The Center for Xizang Chinese (Tibetan) Medicine Resource, Joint Laboratory for Tibetan Materia Medica Resource Scientific Protection and Utilization, Tibetan Medical Research Center of Tibet, Tibet Agriculture and Animal Husbandry University, Nyingchi, Tibet, PR China; Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, GDMU-TAAHC Biomedical and Health Joint R&D Center, Department of Precision Laboratory, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China; Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, PR China.
| |
Collapse
|
8
|
He B, Shao B, Cheng C, Ye Z, Yang Y, Fan B, Xia H, Wu H, Liu Q, Zhang J. miR-21-Mediated Endothelial Senescence and Dysfunction Are Involved in Cigarette Smoke-Induced Pulmonary Hypertension through Activation of PI3K/AKT/mTOR Signaling. TOXICS 2024; 12:396. [PMID: 38922076 PMCID: PMC11209295 DOI: 10.3390/toxics12060396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/27/2024]
Abstract
Smoking is a pathogenic factor for pulmonary hypertension (PH). Our previous study showed that serum miR-21 levels are elevated in smokers. miR-21 is considered as engaged in the PH process; however, its mechanisms remain unclear. In this investigation, we found that in the lung tissue of smoking-induced PH patients, the levels of miR-21 and aging markers (p21 and p16) were upregulated, and the function of pulmonary vascular endothelial cells was also impaired. Exposure of mice to cigarette smoke (CS) for four months caused similar changes in lung tissues and increased pulmonary arterial pressure, which were attenuated by knockout of miR-21. Further, human umbilical vein endothelial cells (HUVECs) exposed to cigarette smoke extract (CSE) revealed upregulation of miR-21 levels, depression of PTEN, activation of PI3K/AKT/mTOR signaling, an increase in senescence indexes, and enhanced dysfunction. Inhibiting miR-21 overexpression reversed the PTEN-mTOR signaling pathway and prevented senescence and dysfunction of HUVECs. In sum, our data indicate that miR-21-mediated endothelial senescence and dysfunction are involved in CS-induced PH through the activation of PI3K/AKT/mTOR signaling, which suggests that selective miR-21 inhibition offers the potential to attenuate PH.
Collapse
Affiliation(s)
- Bin He
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (B.H.); (H.W.)
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Binxia Shao
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
- Department of Emergency, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School of Nanjing University, Nanjing 210008, China
| | - Cheng Cheng
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Zitong Ye
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Yi Yang
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Bowen Fan
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Haibo Xia
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Hao Wu
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (B.H.); (H.W.)
| | - Qizhan Liu
- The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; (B.S.); (C.C.); (Z.Y.); (Y.Y.); (B.F.); (H.X.)
| | - Jinsong Zhang
- Department of Emergency, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; (B.H.); (H.W.)
| |
Collapse
|
9
|
Luo Y, Qi X, Zhang Z, Zhang J, Li B, Shu T, Li X, Hu H, Li J, Tang Q, Zhou Y, Wang M, Fan T, Guo W, Liu Y, Zhang J, Pang J, Yang P, Gao R, Chen W, Yan C, Xing Y, Du W, Wang J, Wang C. Inactivation of Malic Enzyme 1 in Endothelial Cells Alleviates Pulmonary Hypertension. Circulation 2024; 149:1354-1371. [PMID: 38314588 DOI: 10.1161/circulationaha.123.067579] [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: 10/14/2023] [Accepted: 01/10/2024] [Indexed: 02/06/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive cardiopulmonary disease with a high mortality rate. Although growing evidence has revealed the importance of dysregulated energetic metabolism in the pathogenesis of PH, the underlying cellular and molecular mechanisms are not fully understood. In this study, we focused on ME1 (malic enzyme 1), a key enzyme linking glycolysis to the tricarboxylic acid cycle. We aimed to determine the role and mechanistic action of ME1 in PH. METHODS Global and endothelial-specific ME1 knockout mice were used to investigate the role of ME1 in hypoxia- and SU5416/hypoxia (SuHx)-induced PH. Small hairpin RNA and ME1 enzymatic inhibitor (ME1*) were used to study the mechanism of ME1 in pulmonary artery endothelial cells. Downstream key metabolic pathways and mediators of ME1 were identified by metabolomics analysis in vivo and ME1-mediated energetic alterations were examined by Seahorse metabolic analysis in vitro. The pharmacological effect of ME1* on PH treatment was evaluated in PH animal models induced by SuHx. RESULTS We found that ME1 protein level and enzymatic activity were highly elevated in lung tissues of patients and mice with PH, primarily in vascular endothelial cells. Global knockout of ME1 protected mice from developing hypoxia- or SuHx-induced PH. Endothelial-specific ME1 deletion similarly attenuated pulmonary vascular remodeling and PH development in mice, suggesting a critical role of endothelial ME1 in PH. Mechanistic studies revealed that ME1 inhibition promoted downstream adenosine production and activated A2AR-mediated adenosine signaling, which leads to an increase in nitric oxide generation and a decrease in proinflammatory molecule expression in endothelial cells. ME1 inhibition activated adenosine production in an ATP-dependent manner through regulating malate-aspartate NADH (nicotinamide adenine dinucleotide plus hydrogen) shuttle and thereby balancing oxidative phosphorylation and glycolysis. Pharmacological inactivation of ME1 attenuated the progression of PH in both preventive and therapeutic settings by promoting adenosine production in vivo. CONCLUSIONS Our findings indicate that ME1 upregulation in endothelial cells plays a causative role in PH development by negatively regulating adenosine production and subsequently dysregulating endothelial functions. Our findings also suggest that ME1 may represent as a novel pharmacological target for upregulating protective adenosine signaling in PH therapy.
Collapse
Affiliation(s)
- Ya Luo
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
- Department of Pulmonary and Critical Care Medicine, Xinqiao Hospital, Third Military Medical University, Chongqing, China (Y.L.)
| | - Xianmei Qi
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Zhenxi Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases (Z.Z., W.D.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jiawei Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Bolun Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ting Shu
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Xiaona Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Huiyuan Hu
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jinqiu Li
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Qihao Tang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Yitian Zhou
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Mingyao Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China (M.W., C.W.)
| | - Tianfei Fan
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenjun Guo
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ying Liu
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jin Zhang
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China (J.Z.)
| | - Junling Pang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Peiran Yang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Ran Gao
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenhui Chen
- Department of Lung Transplantation, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China (W.C.)
| | - Chen Yan
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY (C.Y.)
| | - Yanjiang Xing
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Wenjing Du
- State Key Laboratory of Common Mechanism Research for Major Diseases (Z.Z., W.D.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Jing Wang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
| | - Chen Wang
- State Key Laboratory of Respiratory Health and Multimorbidity (Y.L., X.Q., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., J.P., P.Y., Y.X., J.W., C.W.)
- Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China (Y.L., X.Q., Z.Z., J.Z., B.L., T.S., X.L., H.H., J.L., Q.T., Y.Z., T.F., W.G., Y.L., J.P., P.Y., R.G., Y.X., W.D., J.W., C.W.)
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing, China (M.W., C.W.)
- Chinese Academy of Engineering, Beijing, China (C.W.)
| |
Collapse
|
10
|
Rao RJ, Chan SY. Mediating Metabolism: Inhibition of Malic Enzyme 1 (ME1) Restores Endothelial Bioenergetics and Adenosine Signaling in Pulmonary Hypertension. Circulation 2024; 149:1372-1374. [PMID: 38648276 PMCID: PMC11045165 DOI: 10.1161/circulationaha.124.068738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Affiliation(s)
- Rashmi J. Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Zhang J, Xu C, Tang X, Sun S, Liu S, Yang L, Chen Y, Yang Q, Wei TYW, Wu X, Wang J, Wang C, Yan X, Yang L, Niu Y, Gou D, Shyy JYJ, Liu B. Endothelium-specific SIRT7 targeting ameliorates pulmonary hypertension through Krüpple-like factor 4 deacetylation. Cardiovasc Res 2024; 120:403-416. [PMID: 38198357 PMCID: PMC10981524 DOI: 10.1093/cvr/cvae011] [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: 05/09/2023] [Revised: 11/11/2023] [Accepted: 11/29/2023] [Indexed: 01/12/2024] Open
Abstract
AIMS Pulmonary hypertension (PH) is a pulmonary vascular disease characterized by a high mortality rate. Pulmonary arterial endothelium cells (PAECs) serve as a primary sensor of various environmental cues, such as shear stress and hypoxia, but PAEC dysfunction may trigger vascular remodelling during the onset of PH. This study aimed to illustrate the role of Sirtuin 7 (SIRT7) in endothelial dysfunction during PH and explore the potential therapeutic strategy for PH. METHODS AND RESULTS SIRT7 levels were measured in human and murine experimental PH samples. Bioinformatic analysis, immunoprecipitation, and deacetylation assay were used to identify the association between SIRT7 and Krüpple-like factor 4 (KLF4), a key transcription factor essential for endothelial cell (EC) homeostasis. Sugen5416 + hypoxia (SuHx)-induced PH mouse models and cell cultures were used for the study of the therapeutic effect of SIRT7 for PH. SIRT7 level was significantly reduced in lung tissues and PAECs from PH patients and the SuHx-induced PH mouse model as compared with healthy controls. Pulmonary endothelium-specific depletion of Sirt7 increased right ventricular systolic pressure and exacerbated right ventricular hypertrophy in the SuHx-induced PH model. At the molecular level, we identified KLF4 as a downstream target of SIRT7, which deacetylated KLF4 at K228 and inhibited the ubiquitination-proteasome degradation. Thus, the SIRT7/KLF4 axis maintained PAEC homeostasis by regulating proliferation, migration, and tube formation. PAEC dysfunction was reversed by adeno-associated virus type 1 vector-mediated endothelial overexpression of Sirt7 or supplementation with nicotinamide adenine dinucleotide (NAD)+ intermediate nicotinamide riboside which activated Sirt7; both approaches successfully reversed PH phenotypes. CONCLUSION The SIRT7/KLF4 axis ensures PAEC homeostasis, and pulmonary endothelium-specific SIRT7 targeting might constitute a PH therapeutic strategy.
Collapse
Affiliation(s)
- Jin Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| | - Chenzhong Xu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Marshall Laboratory of Biomedical Engineering, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen 518060, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| | - Xiaolong Tang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| | - Shimin Sun
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| | - Siqi Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| | - Langmei Yang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| | - Yuqin Chen
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Qifeng Yang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Tong-You Wade Wei
- Division of Cardiology, Department of Medicine, University of California, San Diego 9500 Gilman Dr, La Jolla, CA 92023, USA
| | - Xiaojing Wu
- Cardiovascular Department of Shenzhen University General Hospital, Shenzhen 518055, China
| | - Jian Wang
- State Key Laboratory of Respiratory Diseases, National Center for Respiratory Medicine, Guangdong Key Laboratory of Vascular Diseases, National Clinical Research Center for Respiratory Diseases, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, China
| | - Chen Wang
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xiaosong Yan
- Department of Pathology, The Affiliated Children’s Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710003, China
| | - Lei Yang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, 1066 Xueyuan Blvd, Nanshan District, Shenzhen, 518060, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, 1066 Xueyuan Blvd, Nanshan District, Shenzhen, 518060, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Vascular Disease Research Center, College of Life Sciences and Oceanography, Shenzhen University, 1066 Xueyuan Blvd, Nanshan District, Shenzhen, 518060, China
| | - John Y J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego 9500 Gilman Dr, La Jolla, CA 92023, USA
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), International Cancer Center, School of Basic Medical Sciences, Shenzhen University Medical School, 1066 Xueyuan Blvd, Nanshan District, Shenzhen 518055, China
| |
Collapse
|
12
|
Piper B, Bogamuwa S, Hossain T, Farkas D, Rosas L, Green AC, Newcomb G, Sun N, Ovando-Ricardez JA, 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. J Clin Invest 2024; 134:e169441. [PMID: 38015641 PMCID: PMC10836802 DOI: 10.1172/jci169441] [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: 02/07/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating and progressive disease with limited treatment options. Endothelial dysfunction plays a central role in the 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 patients with PAH. Endothelial haploinsufficiency of RAB7 caused spontaneous pulmonary hypertension (PH) in mice. Silencing of RAB7 in ECs induced broad changes in gene expression revealed via RNA-Seq, and RAB7-silenced ECs showed impaired angiogenesis and expansion of a senescent cell fraction, combined with impaired endolysosomal trafficking and degradation, suggesting inhibition of autophagy at the predegradation level. Furthermore, mitochondrial membrane potential and oxidative phosphorylation were decreased, and glycolysis was enhanced. Treatment with the RAB7 activator ML-098 reduced established PH in rats with chronic hypoxia/SU5416. In conclusion, we demonstrate for the first time to our knowledge the fundamental impairment of EC function by loss of RAB7, causing PH, and show RAB7 activation to be a potential therapeutic strategy in a preclinical model of PH.
Collapse
Affiliation(s)
- Bryce Piper
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Srimathi Bogamuwa
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | | | - Daniela Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Lorena Rosas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | | | - Geoffrey Newcomb
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Nuo Sun
- Davis Heart and Lung Research Institute
- Department of Cell Biology and Physiology, The Ohio State University (OSU), Columbus, Ohio, USA
| | - Jose A. Ovando-Ricardez
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Jeffrey C. Horowitz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Aneel R. Bhagwani
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
- Department of Physiology, Ziauddin University, Karachi, Pakistan
| | - Hu Yang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri, USA
| | - Tatiana V. Kudryashova
- University of Pittsburgh, Heart, Blood, and Vascular Medicine Institute, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Ana L. Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Pearlly Yan
- Division of Hematology, Department of Internal Medicine and The James Cancer Center, OSU, Columbus, Ohio, USA
| | - Rama K. Mallampalli
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| | - Elena A. Goncharova
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California Davis, Davis, California, USA
| | - David M. Eckmann
- Department of Anesthesiology, and
- Center for Medical and Engineering Innovation, OSU, Columbus, Ohio, USA
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine
- Davis Heart and Lung Research Institute
| |
Collapse
|
13
|
Tai YY, Yu Q, Tang Y, Sun W, Kelly NJ, Okawa S, Zhao J, Schwantes-An TH, Lacoux C, Torrino S, Aaraj YA, Khoury WE, Negi V, Liu M, Corey CG, Belmonte F, Vargas SO, Schwartz B, Bhat B, Chau BN, Karnes JH, Satoh T, Barndt RJ, Wu H, Parikh VN, Wang J, Zhang Y, McNamara D, Li G, Speyer G, Wang B, Shiva S, Kaufman B, Kim S, Gomez D, Mari B, Cho MH, Boueiz A, Pauciulo MW, Southgate L, Trembath RC, Sitbon O, Humbert M, Graf S, Morrell NW, Rhodes CJ, Wilkins MR, Nouraie M, Nichols WC, Desai AA, Bertero T, Chan SY. Allele-specific control of rodent and human lncRNA KMT2E-AS1 promotes hypoxic endothelial pathology in pulmonary hypertension. Sci Transl Med 2024; 16:eadd2029. [PMID: 38198571 PMCID: PMC10947529 DOI: 10.1126/scitranslmed.add2029] [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: 05/26/2022] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Hypoxic reprogramming of vasculature relies on genetic, epigenetic, and metabolic circuitry, but the control points are unknown. In pulmonary arterial hypertension (PAH), a disease driven by hypoxia inducible factor (HIF)-dependent vascular dysfunction, HIF-2α promoted expression of neighboring genes, long noncoding RNA (lncRNA) histone lysine N-methyltransferase 2E-antisense 1 (KMT2E-AS1) and histone lysine N-methyltransferase 2E (KMT2E). KMT2E-AS1 stabilized KMT2E protein to increase epigenetic histone 3 lysine 4 trimethylation (H3K4me3), driving HIF-2α-dependent metabolic and pathogenic endothelial activity. This lncRNA axis also increased HIF-2α expression across epigenetic, transcriptional, and posttranscriptional contexts, thus promoting a positive feedback loop to further augment HIF-2α activity. We identified a genetic association between rs73184087, a single-nucleotide variant (SNV) within a KMT2E intron, and disease risk in PAH discovery and replication patient cohorts and in a global meta-analysis. This SNV displayed allele (G)-specific association with HIF-2α, engaged in long-range chromatin interactions, and induced the lncRNA-KMT2E tandem in hypoxic (G/G) cells. In vivo, KMT2E-AS1 deficiency protected against PAH in mice, as did pharmacologic inhibition of histone methylation in rats. Conversely, forced lncRNA expression promoted more severe PH. Thus, the KMT2E-AS1/KMT2E pair orchestrates across convergent multi-ome landscapes to mediate HIF-2α pathobiology and represents a key clinical target in pulmonary hypertension.
Collapse
Affiliation(s)
- Yi Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Qiujun Yu
- Cardiovascular Division, Department Of Internal Medicine, Washington University School of Medicine, St. louis, Mo 63110, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Neil J. Kelly
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Va Medical Center, Pittsburgh, PA 15240, USA
| | - Satoshi Okawa
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15219, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Tae-Hwi Schwantes-An
- Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, In 46202, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, In 46202, USA
| | - Caroline Lacoux
- Université côte d’Azur, CNRS, IPMC, IHU RespiERA, Sophia-Antipolis, 06903, France
| | - Stephanie Torrino
- Université côte d’Azur, CNRS, IPMC, IHU RespiERA, Sophia-Antipolis, 06903, France
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Vinny Negi
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Mingjun Liu
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Catherine G. Corey
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Pediatrics, University of Pittsburgh Medical center children’s hospital, Pittsburgh, PA 15224, USA
| | - Frances Belmonte
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Sara O. Vargas
- Department of Pathology, Boston Children’s Hospital, Boston, MA 02115, USA
| | | | - Bal Bhat
- Translate Bio, Lexington, MA 02421, USA
| | | | - Jason H. Karnes
- Division of Pharmacogenomics, College of Pharmacy, University of Arizona College of Medicine, Tucson, AZ 85721, USA
| | - Taijyu Satoh
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, 980–8575, Japan
| | - Robert J. Barndt
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Haodi Wu
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Victoria N. Parikh
- Stanford Center for Inherited Cardiovascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jianrong Wang
- Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Yingze Zhang
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Dennis McNamara
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Gang Li
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Aging Institute, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Gil Speyer
- Research Computing, Arizona State University, Tempe, AZ 85281, USA
| | - Bing Wang
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Sruti Shiva
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Pharmacology and chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Brett Kaufman
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Seungchan Kim
- Center for Computational Systems Biology, Department of Electrical and Computer Engineering, Roy G. Perry college of Engineering, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Delphine Gomez
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Bernard Mari
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, In 46202, USA
| | - Michael H. Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Adel Boueiz
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael W. Pauciulo
- Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Laura Southgate
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, WC2R 2lS, UK
- Molecular and Clinical Sciences Research Institute, St George’s University of London, London, SW17 0RE, UK
| | - Richard C. Trembath
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College London, London, WC2R 2lS, UK
| | - Olivier Sitbon
- Université Paris–Saclay, INSERM, Assistance Publique Hôpitaux de Paris, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin Bicêtre, 94270, France
| | - Marc Humbert
- Université Paris–Saclay, INSERM, Assistance Publique Hôpitaux de Paris, Service de Pneumologie et Soins Intensifs Respiratoires, Hôpital Bicêtre, Le Kremlin Bicêtre, 94270, France
| | - Stefan Graf
- Department of Medicine, University of Cambridge, Cambridge, CB2 1TN, UK
- NIHR Bioresource for Translational Research, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Department of Haematology, University of Cambridge, NHS Blood and Transplant, Long Road, Cambridge, CB2 2PT, UK
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge, Cambridge, CB2 1TN, UK
- Centessa Pharmaceuticals, Altrincham, Cheshire, WA14 2DT, UK
| | | | - Martin R. Wilkins
- National Heart and Lung Institute, Imperial College London, London, SW3 6lY, UK
| | - Mehdi Nouraie
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - William C. Nichols
- Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Ankit A. Desai
- Division of Cardiology, Department of Medicine, Indiana University School of Medicine, Indianapolis, In 46202, USA
| | - Thomas Bertero
- Université côte d’Azur, CNRS, IPMC, IHU RespiERA, Sophia-Antipolis, 06903, France
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Division of cardiology, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
- Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
14
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
15
|
Gao J, Huang C, Kong L, Zhou W, Sun M, Wei T, Shen W. SIRT3 Regulates Clearance of Apoptotic Cardiomyocytes by Deacetylating Frataxin. Circ Res 2023; 133:631-647. [PMID: 37646156 PMCID: PMC10498872 DOI: 10.1161/circresaha.123.323160] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Efferocytosis is an activity of macrophages that is pivotal for the resolution of inflammation in hypertension. The precise mechanism by which macrophages coordinate efferocytosis and internalize apoptotic cardiomyocytes remains unknown. The aim of this study was to determine whether SIRT3 (sirtuin-3) is required for both apoptotic cardiomyocyte engulfment and anti-inflammatory responses during efferocytosis. METHODS We generated myeloid SIRT3 knockout mice and FXN (frataxin) knock-in mice carrying an acetylation-defective lysine to arginine K189R mutation (FXNK189R). The mice were given Ang II (angiotensin II) infusion for 7 days. We analyzed cardiac macrophages' mitochondrial iron levels, efferocytosis activity, and phenotype both in vivo and in vitro. RESULTS We showed that SIRT3 deficiency exacerbated Ang II-induced downregulation of the efferocytosis receptor MerTK (c-Mer tyrosine kinase) and proinflammatory cytokine production, accompanied by disrupted mitochondrial iron homeostasis in cardiac macrophages. Quantitative acetylome analysis revealed that SIRT3 deacetylated FXN at lysine 189. Ang II attenuated SIRT3 activity and enhanced the acetylation level of FXNK189. Acetylated FXN further reduced the synthesis of ISCs (iron-sulfur clusters), resulting in mitochondrial iron accumulation. Phagocytic internalization of apoptotic cardiomyocytes increased myoglobin content, and derived iron ions promoted mitochondrial iron overload and lipid peroxidation. An iron chelator deferoxamine improved the levels of MerTK and efferocytosis, thereby attenuating proinflammatory macrophage activation. FXNK189R mice showed improved macrophage efferocytosis, reduced cardiac inflammation, and suppressed cardiac fibrosis. CONCLUSIONS The SIRT3-FXN axis has the potential to resolve cardiac inflammation by increasing macrophage efferocytosis and anti-inflammatory activities.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital (J.G., C.H., L.K., T.W., W.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Chenglin Huang
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital (J.G., C.H., L.K., T.W., W.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Linghui Kong
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital (J.G., C.H., L.K., T.W., W.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Wugang Zhou
- Department of Emergency, Shanghai Ninth People’s Hospital (W.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Mengwei Sun
- Department of Emergency, Shanghai Ninth People’s Hospital (W.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Tong Wei
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital (J.G., C.H., L.K., T.W., W.S.), Shanghai Jiao Tong University School of Medicine, China
| | - Weili Shen
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital (J.G., C.H., L.K., T.W., W.S.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
16
|
Fujiwara T, Takeda N, Hara H, Ishii S, Numata G, Tokiwa H, Katoh M, Maemura S, Suzuki T, Takiguchi H, Yanase T, Kubota Y, Nomura S, Hatano M, Ueda K, Harada M, Toko H, Takimoto E, Akazawa H, Morita H, Nishimura S, Komuro I. PGC-1α-mediated angiogenesis prevents pulmonary hypertension in mice. JCI Insight 2023; 8:e162632. [PMID: 37681410 PMCID: PMC10544206 DOI: 10.1172/jci.insight.162632] [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: 06/13/2022] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Pulmonary hypertension (PH) is a life-threatening disease characterized by a progressive narrowing of pulmonary arterioles. Although VEGF is highly expressed in lung of patients with PH and in animal PH models, the involvement of angiogenesis remains elusive. To clarify the pathophysiological function of angiogenesis in PH, we compared the angiogenic response in hypoxia (Hx) and SU5416 (a VEGFR2 inhibitor) plus Hx (SuHx) mouse PH models using 3D imaging. The 3D imaging analysis revealed an angiogenic response in the lung of the Hx-PH, but not of the severer SuHx-PH model. Selective VEGFR2 inhibition with cabozantinib plus Hx in mice also suppressed angiogenic response and exacerbated Hx-PH to the same extent as SuHx. Expression of endothelial proliferator-activated receptor γ coactivator 1α (PGC-1α) increased along with angiogenesis in lung of Hx-PH but not SuHx mice. In pulmonary endothelial cell-specific Ppargc1a-KO mice, the Hx-induced angiogenesis was suppressed, and PH was exacerbated along with increased oxidative stress, cellular senescence, and DNA damage. By contrast, treatment with baicalin, a flavonoid enhancing PGC-1α activity in endothelial cells, ameliorated Hx-PH with increased Vegfa expression and angiogenesis. Pulmonary endothelial PGC-1α-mediated angiogenesis is essential for adaptive responses to Hx and might represent a potential therapeutic target for PH.
Collapse
Affiliation(s)
- Takayuki Fujiwara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Computational Diagnostic Radiology and Preventive Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Hironori Hara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Satoshi Ishii
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Genri Numata
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Tokiwa
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Manami Katoh
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Sonoko Maemura
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Takaaki Suzuki
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Takiguchi
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Tomonobu Yanase
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Therapeutic Strategy for Heart Failure, and
| | - Masaru Hatano
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Kazutaka Ueda
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Mutsuo Harada
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
- Department of Advanced Clinical Science and Therapeutics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Haruhiro Toko
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Satoshi Nishimura
- Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
17
|
Culley MK, Rao RJ, Mehta M, Zhao J, El Khoury W, Harvey LD, Perk D, Tai YY, Tang Y, Shiva S, Rabinovitch M, Gu M, Bertero T, Chan SY. Frataxin deficiency disrupts mitochondrial respiration and pulmonary endothelial cell function. Vascul Pharmacol 2023; 151:107181. [PMID: 37164245 PMCID: PMC10524929 DOI: 10.1016/j.vph.2023.107181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 04/19/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Deficiency of iron‑sulfur (FeS) clusters promotes metabolic rewiring of the endothelium and the development of pulmonary hypertension (PH) in vivo. Joining a growing number of FeS biogenesis proteins critical to pulmonary endothelial function, recent data highlighted that frataxin (FXN) reduction drives Fe-S-dependent genotoxic stress and senescence across multiple types of pulmonary vascular disease. Trinucleotide repeat mutations in the FXN gene cause Friedreich's ataxia, a disease characterized by cardiomyopathy and neurodegeneration. These tissue-specific phenotypes have historically been attributed to mitochondrial reprogramming and oxidative stress. Whether FXN coordinates both nuclear and mitochondrial processes in the endothelium is unknown. Here, we aim to identify the mitochondria-specific effects of FXN deficiency in the endothelium that predispose to pulmonary hypertension. Our data highlight an Fe-S-driven metabolic shift separate from previously described replication stress whereby FXN knockdown diminished mitochondrial respiration and increased glycolysis and oxidative species production. In turn, FXN-deficient endothelial cells had increased vasoconstrictor production (EDN1) and decreased nitric oxide synthase expression (NOS3). These data were observed in primary pulmonary endothelial cells after pharmacologic inhibition of FXN, mice carrying a genetic endothelial deletion of FXN, and inducible pluripotent stem cell-derived endothelial cells from patients with FXN mutations. Altogether, this study indicates FXN is an upstream driver of pathologic aberrations in metabolism and genomic stability. Moreover, our study highlights FXN-specific vasoconstriction in vivo, prompting future studies to investigate available and novel PH therapies in contexts of FXN deficiency.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Rashmi J Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Monica Mehta
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lloyd D Harvey
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Dror Perk
- Medical Scientist Training Program, Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yi Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sruti Shiva
- Department of Pharmacology and Chemical Biology, Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, USA
| | - Marlene Rabinovitch
- Stanford Children's Health Betty Irene Moore Children's Heart Center, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mingxia Gu
- Perinatal Institute, Division of Pulmonary Biology Center for Stem Cell and Organoid Medicine, CuSTOM, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Thomas Bertero
- Université Côte d'Azur, CNRS, UMR7275, IPMC, Valbonne, France
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Santos AK, Scalzo S, de Souza RTV, Santana PHG, Marques BL, Oliveira LF, Filho DM, Kihara AH, da Costa Santiago H, Parreira RC, Birbrair A, Ulrich H, Resende RR. Strategic use of organoids and organs-on-chip as biomimetic tools. Semin Cell Dev Biol 2023; 144:3-10. [PMID: 36192310 DOI: 10.1016/j.semcdb.2022.09.010] [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: 06/14/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/30/2022]
Abstract
Organoid development and organ-on-a-chip are technologies based on differentiating stem cells, forming 3D multicellular structures resembling organs and tissues in vivo. Hence, both can be strategically used for disease modeling, drug screening, and host-pathogen studies. In this context, this review highlights the significant advancements in the area, providing technical approaches to organoids and organ-on-a-chip that best imitate in vivo physiology.
Collapse
Affiliation(s)
- Anderson K Santos
- Department of Pediatrics, Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Sérgio Scalzo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, GO, Brazil
| | - Lucas F Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal do Triângulo Mineiro, Uberaba, MG, Brazil
| | - Daniel M Filho
- Departamento de Fisiologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Alexandre Hiroaki Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Helton da Costa Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Alexander Birbrair
- Departmento de Patologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA; Department of Radiology, Columbia University Medical Center, New York, NY, USA
| | - Henning Ulrich
- Departmento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinópolis, Brazil.
| |
Collapse
|
19
|
Immanuel J, Yun S. Vascular Inflammatory Diseases and Endothelial Phenotypes. Cells 2023; 12:1640. [PMID: 37371110 PMCID: PMC10297687 DOI: 10.3390/cells12121640] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The physiological functions of endothelial cells control vascular tone, permeability, inflammation, and angiogenesis, which significantly help to maintain a healthy vascular system. Several cardiovascular diseases are characterized by endothelial cell activation or dysfunction triggered by external stimuli such as disturbed flow, hypoxia, growth factors, and cytokines in response to high levels of low-density lipoprotein and cholesterol, hypertension, diabetes, aging, drugs, and smoking. Increasing evidence suggests that uncontrolled proinflammatory signaling and further alteration in endothelial cell phenotypes such as barrier disruption, increased permeability, endothelial to mesenchymal transition (EndMT), and metabolic reprogramming further induce vascular diseases, and multiple studies are focusing on finding the pathways and mechanisms involved in it. This review highlights the main proinflammatory stimuli and their effects on endothelial cell function. In order to provide a rational direction for future research, we also compiled the most recent data regarding the impact of endothelial cell dysfunction on vascular diseases and potential targets that impede the pathogenic process.
Collapse
Affiliation(s)
| | - Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae-si 50834, Republic of Korea;
| |
Collapse
|
20
|
Chen PY, Qin L, Simons M. TGFβ signaling pathways in human health and disease. Front Mol Biosci 2023; 10:1113061. [PMID: 37325472 PMCID: PMC10267471 DOI: 10.3389/fmolb.2023.1113061] [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: 12/01/2022] [Accepted: 04/27/2023] [Indexed: 06/17/2023] Open
Abstract
Transforming growth factor beta (TGFβ) is named for the function it was originally discovered to perform-transformation of normal cells into aggressively growing malignant cells. It became apparent after more than 30 years of research, however, that TGFβ is a multifaceted molecule with a myriad of different activities. TGFβs are widely expressed with almost every cell in the human body producing one or another TGFβ family member and expressing its receptors. Importantly, specific effects of this growth factor family differ in different cell types and under different physiologic and pathologic conditions. One of the more important and critical TGFβ activities is the regulation of cell fate, especially in the vasculature, that will be the focus of this review.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
21
|
Riou M, Enache I, Sauer F, Charles AL, Geny B. Targeting Mitochondrial Metabolic Dysfunction in Pulmonary Hypertension: Toward New Therapeutic Approaches? Int J Mol Sci 2023; 24:ijms24119572. [PMID: 37298522 DOI: 10.3390/ijms24119572] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare disease characterized by pulmonary vascular remodeling leading to right heart failure and death. To date, despite the three therapeutic approaches targeting the three major endothelial dysfunction pathways based on the prostacyclin, nitric oxide/cyclic guanosine monophosphate, and endothelin pathways, PAH remains a serious disease. As such, new targets and therapeutic agents are needed. Mitochondrial metabolic dysfunction is one of the mechanisms involved in PAH pathogenesis in part through the induction of a Warburg metabolic state of enhanced glycolysis but also through the upregulation of glutaminolysis, tricarboxylic cycle and electron transport chain dysfunction, dysregulation of fatty acid oxidation or mitochondrial dynamics alterations. The aim of this review is to shed light on the main mitochondrial metabolic pathways involved in PAH and to provide an update on the resulting interesting potential therapeutic perspectives.
Collapse
Affiliation(s)
- Marianne Riou
- Translational Medicine Federation of Strasbourg (FMTS), CRBS, University of Strasbourg, Team 3072 "Mitochondria, Oxidative Stress and Muscle Protection", 1 Rue Eugène Boeckel, CS 60026, CEDEX 67084 Strasbourg, France
- Physiology and Functional Exploration Unit, University Hospital of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France
| | - Irina Enache
- Translational Medicine Federation of Strasbourg (FMTS), CRBS, University of Strasbourg, Team 3072 "Mitochondria, Oxidative Stress and Muscle Protection", 1 Rue Eugène Boeckel, CS 60026, CEDEX 67084 Strasbourg, France
- Physiology and Functional Exploration Unit, University Hospital of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France
| | - François Sauer
- Translational Medicine Federation of Strasbourg (FMTS), CRBS, University of Strasbourg, Team 3072 "Mitochondria, Oxidative Stress and Muscle Protection", 1 Rue Eugène Boeckel, CS 60026, CEDEX 67084 Strasbourg, France
- Cardiology Unit, University Hospital of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France
| | - Anne-Laure Charles
- Translational Medicine Federation of Strasbourg (FMTS), CRBS, University of Strasbourg, Team 3072 "Mitochondria, Oxidative Stress and Muscle Protection", 1 Rue Eugène Boeckel, CS 60026, CEDEX 67084 Strasbourg, France
| | - Bernard Geny
- Translational Medicine Federation of Strasbourg (FMTS), CRBS, University of Strasbourg, Team 3072 "Mitochondria, Oxidative Stress and Muscle Protection", 1 Rue Eugène Boeckel, CS 60026, CEDEX 67084 Strasbourg, France
- Physiology and Functional Exploration Unit, University Hospital of Strasbourg, 1 Place de l'Hôpital, CEDEX 67091 Strasbourg, France
| |
Collapse
|
22
|
Ramadhiani R, Ikeda K, Miyagawa K, Ryanto GRT, Tamada N, Suzuki Y, Kirita Y, Matoba S, Hirata KI, Emoto N. Endothelial cell senescence exacerbates pulmonary hypertension by inducing juxtacrine Notch signaling in smooth muscle cells. iScience 2023; 26:106662. [PMID: 37192975 PMCID: PMC10182325 DOI: 10.1016/j.isci.2023.106662] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 03/17/2023] [Accepted: 04/06/2023] [Indexed: 05/18/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease characterized by a progressive increase in pulmonary artery pressure caused by pathological pulmonary artery remodeling. Here, we demonstrate that endothelial cell (EC) senescence plays a negative role in pulmonary hypertension via juxtacrine interaction with smooth muscle cells (SMCs). By using EC-specific progeroid mice, we discovered that EC progeria deteriorated vascular remodeling in the lungs, and exacerbated pulmonary hypertension in mice. Mechanistically, senescent ECs overexpressed Notch ligands, which resulted in increased Notch signaling and activated proliferation and migration capacities in neighboring SMCs. Pharmacological inhibition of Notch signaling reduced the effects of senescent ECs on SMCs functions in vitro, and improved the worsened pulmonary hypertension in EC-specific progeroid mice in vivo. Our findings show that EC senescence is a critical disease-modifying factor in PAH and that EC-mediated Notch signaling is a pharmacotherapeutic target for the treatment of PAH, particularly in the elderly.
Collapse
Affiliation(s)
- Risa Ramadhiani
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Koji Ikeda
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Department of Epidemiology for Longevity and Regional Health, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Kazuya Miyagawa
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Gusty Rizky Tough Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Naoki Tamada
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Yoko Suzuki
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
| | - Yuhei Kirita
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Satoaki Matoba
- Department of Cardiology and Nephrology, Kyoto Prefectural University of Medicine, 465 Kajii, Kawaramachi-Hirokoji, Kamigyou, Kyoto 6028566, Japan
| | - Ken-ichi Hirata
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada, Kobe 658-8558, Japan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe 6500017, Japan
| |
Collapse
|
23
|
Suda M, Paul KH, Minamino T, Miller JD, Lerman A, Ellison-Hughes GM, Tchkonia T, Kirkland JL. Senescent Cells: A Therapeutic Target in Cardiovascular Diseases. Cells 2023; 12:1296. [PMID: 37174697 PMCID: PMC10177324 DOI: 10.3390/cells12091296] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Senescent cell accumulation has been observed in age-associated diseases including cardiovascular diseases. Senescent cells lack proliferative capacity and secrete senescence-associated secretory phenotype (SASP) factors that may cause or worsen many cardiovascular diseases. Therapies targeting senescent cells, especially senolytic drugs that selectively induce senescent cell removal, have been shown to delay, prevent, alleviate, or treat multiple age-associated diseases in preclinical models. Some senolytic clinical trials have already been completed or are underway for a number of diseases and geriatric syndromes. Understanding how cellular senescence affects the various cell types in the cardiovascular system, such as endothelial cells, vascular smooth muscle cells, fibroblasts, immune cells, progenitor cells, and cardiomyocytes, is important to facilitate translation of senotherapeutics into clinical interventions. This review highlights: (1) the characteristics of senescent cells and their involvement in cardiovascular diseases, focusing on the aforementioned cardiovascular cell types, (2) evidence about senolytic drugs and other senotherapeutics, and (3) the future path and clinical potential of senotherapeutics for cardiovascular diseases.
Collapse
Affiliation(s)
- Masayoshi Suda
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Karl H. Paul
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo 100-0004, Japan
| | - Jordan D. Miller
- Division of Cardiovascular Surgery, Mayo Clinic College of Medicine, 200 First St., S.W., Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - Georgina M. Ellison-Hughes
- Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
- Centre for Stem Cells and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences & Medicine, Guy’s Campus, King’s College London, London SE1 1UL, UK
| | - Tamar Tchkonia
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| | - James L. Kirkland
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, 200 First St., S.W., Rochester, MN 55905, USA
| |
Collapse
|
24
|
Hafeez N, Kirillova A, Yue Y, Rao RJ, Kelly NJ, El Khoury W, Al Aaraj Y, Tai Y, Handen A, Tang Y, Jiang D, Wu T, Zhang Y, McNamara D, Kudryashova TV, Goncharova EA, Goncharov D, Bertero T, Nouraie M, Li G, Sun W, Chan SY. Single Nucleotide Polymorphism rs9277336 Controls the Nuclear Alpha Actinin 4-Human Leukocyte Antigen-DPA1 Axis and Pulmonary Endothelial Pathophenotypes in Pulmonary Arterial Hypertension. J Am Heart Assoc 2023; 12:e027894. [PMID: 36974749 PMCID: PMC10122886 DOI: 10.1161/jaha.122.027894] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 02/06/2023] [Indexed: 03/29/2023]
Abstract
Background Pulmonary arterial hypertension (PAH) is a complex, fatal disease where disease severity has been associated with the single nucleotide polymorphism (SNP) rs2856830, located near the human leukocyte antigen DPA1 (HLA-DPA1) gene. We aimed to define the genetic architecture of functional variants associated with PAH disease severity by identifying allele-specific binding transcription factors and downstream targets that control endothelial pathophenotypes and PAH. Methods and Results Electrophoretic mobility shift assays of oligonucleotides containing SNP rs2856830 and 8 SNPs in linkage disequilibrium revealed functional SNPs via allele-imbalanced binding to human pulmonary arterial endothelial cell nuclear proteins. DNA pulldown proteomics identified SNP-binding proteins. SNP genotyping and clinical correlation analysis were performed in 84 patients with PAH at University of Pittsburgh Medical Center and in 679 patients with PAH in the All of Us database. SNP rs9277336 was identified as a functional SNP in linkage disequilibrium (r2>0.8) defined by rs2856830, and the minor allele was associated with decreased hospitalizations and improved cardiac output in patients with PAH, an index of disease severity. SNP pulldown proteomics showed allele-specific binding of nuclear ACTN4 (alpha actinin 4) protein to rs9277336 minor allele. Both ACTN4 and HLA-DPA1 were downregulated in pulmonary endothelium in human patients and rodent models of PAH. Via transcriptomic and phenotypic analyses, knockdown of HLA-DPA1 phenocopied knockdown of ACTN4, both similarly controlling cell structure pathways, immune pathways, and endothelial dysfunction. Conclusions We defined the pathogenic activity of functional SNP rs9277336, entailing the allele-specific binding of ACTN4 and controlling expression of the neighboring HLA-DPA1 gene. Through inflammatory or genetic means, downregulation of this ACTN4-HLA-DPA1 regulatory axis promotes endothelial pathophenotypes, providing a mechanistic explanation for the association between this SNP and PAH outcomes.
Collapse
Affiliation(s)
- Neha Hafeez
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Anna Kirillova
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yunshan Yue
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
- School of MedicineTsinghua UniversityBeijingChina
| | - Rashmi J. Rao
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Neil J. Kelly
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Wadih El Khoury
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yassmin Al Aaraj
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Yi‐Yin Tai
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Adam Handen
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Ying Tang
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Danli Jiang
- The Aging InstituteUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Ting Wu
- The Aging InstituteUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Yingze Zhang
- Division of Pulmonary Allergy and Critical Care Medicine, Department of MedicineUniversity of Pittsburgh Medical CenterPittsburghPA
| | - Dennis McNamara
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Tatiana V. Kudryashova
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal MedicineUniversity of California DavisDavisCA
| | - Elena A. Goncharova
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal MedicineUniversity of California DavisDavisCA
| | - Dmitry Goncharov
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal MedicineUniversity of California DavisDavisCA
| | - Thomas Bertero
- Université Côte d’Azur, CNRS, UMR7275, IPMCValbonneFrance
| | - Mehdi Nouraie
- Division of Pulmonary Allergy and Critical Care Medicine, Department of MedicineUniversity of Pittsburgh Medical CenterPittsburghPA
| | - Gang Li
- The Aging InstituteUniversity of Pittsburgh School of MedicinePittsburghPA
| | - Wei Sun
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| | - Stephen Y. Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, Division of Cardiology, Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPA
| |
Collapse
|
25
|
Sweeney M, Cook SA, Gil J. Therapeutic opportunities for senolysis in cardiovascular disease. FEBS J 2023; 290:1235-1255. [PMID: 35015342 PMCID: PMC10952275 DOI: 10.1111/febs.16351] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/20/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022]
Abstract
Cellular senescence within the cardiovascular system has, until recently, been understudied and unappreciated as a factor in the development of age-related cardiovascular diseases such as heart failure, myocardial infarction and atherosclerosis. This is in part due to challenges with defining senescence within post-mitotic cells such as cardiomyocytes. However, recent evidence has demonstrated senescent-like changes, including a senescence-associated secretory phenotype (SASP), in cardiomyocytes in response to ageing and cell stress. Other replicating cells, including fibroblasts and vascular smooth muscle cells, within the cardiovascular system have also been shown to undergo senescence and contribute to disease pathogenesis. These findings coupled with the emergence of senolytic therapies, to target and eliminate senescent cells, have provided fascinating new avenues for management of several age-related cardiovascular diseases with high prevalence. In this review, we discuss the role of senescent cells within the cardiovascular system and highlight the contribution of senescence cells to common cardiovascular diseases. We discuss the emerging role for senolytics in cardiovascular disease management while highlighting important aspects of senescence biology which must be clarified before the potential of senolytics can be fully realized.
Collapse
Affiliation(s)
- Mark Sweeney
- MRC London Institute of Medical Sciences (LMS)LondonUK
- Institute of Clinical Sciences (ICS)Faculty of MedicineImperial College LondonUK
- Wellcome Trust / National Institute of Health Research 4i Clinical Research FellowLondonUK
| | - Stuart A. Cook
- MRC London Institute of Medical Sciences (LMS)LondonUK
- Institute of Clinical Sciences (ICS)Faculty of MedicineImperial College LondonUK
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS)LondonUK
- Institute of Clinical Sciences (ICS)Faculty of MedicineImperial College LondonUK
| |
Collapse
|
26
|
Born E, Lipskaia L, Breau M, Houssaini A, Beaulieu D, Marcos E, Pierre R, Do Cruzeiro M, Lefevre M, Derumeaux G, Bulavin DV, Delcroix M, Quarck R, Reen V, Gil J, Bernard D, Flaman JM, Adnot S, Abid S. Eliminating Senescent Cells Can Promote Pulmonary Hypertension Development and Progression. Circulation 2023; 147:650-666. [PMID: 36515093 DOI: 10.1161/circulationaha.122.058794] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Senescent cells (SCs) are involved in proliferative disorders, but their role in pulmonary hypertension remains undefined. We investigated SCs in patients with pulmonary arterial hypertension and the role of SCs in animal pulmonary hypertension models. METHODS We investigated senescence (p16, p21) and DNA damage (γ-H2AX, 53BP1) markers in patients with pulmonary arterial hypertension and murine models. We monitored p16 activation by luminescence imaging in p16-luciferase (p16LUC/+) knock-in mice. SC clearance was obtained by a suicide gene (p16 promoter-driven killer gene construct in p16-ATTAC mice), senolytic drugs (ABT263 and cell-permeable FOXO4-p53 interfering peptide [FOXO4-DRI]), and p16 inactivation in p16LUC/LUC mice. We investigated pulmonary hypertension in mice exposed to normoxia, chronic hypoxia, or hypoxia+Sugen, mice overexpressing the serotonin transporter (SM22-5-HTT+), and rats given monocrotaline. RESULTS Patients with pulmonary arterial hypertension compared with controls exhibited high lung p16, p21, and γ-H2AX protein levels, with abundant vascular cells costained for p16, γ-H2AX, and 53BP1. Hypoxia increased thoracic bioluminescence in p16LUC/+ mice. In wild-type mice, hypoxia increased lung levels of senescence and DNA-damage markers, senescence-associated secretory phenotype components, and p16 staining of pulmonary endothelial cells (P-ECs, 30% of lung SCs in normoxia), and pulmonary artery smooth muscle cells. SC elimination by suicide gene or ABT263 increased the right ventricular systolic pressure and hypertrophy index, increased vessel remodeling (higher dividing proliferating cell nuclear antigen-stained vascular cell counts during both normoxia and hypoxia), and markedly decreased lung P-ECs. Pulmonary hemodynamic alterations and lung P-EC loss occurred in older p16LUC/LUC mice, wild-type mice exposed to Sugen or hypoxia+Sugen, and SM22-5-HTT+ mice given either ABT263 or FOXO4-DRI, compared with relevant controls. The severity of monocrotaline-induced pulmonary hypertension in rats was decreased slightly by ABT263 for 1 week but was aggravated at 3 weeks, with loss of P-ECs. CONCLUSIONS Elimination of senescent P-ECs by senolytic interventions may worsen pulmonary hemodynamics. These results invite consideration of the potential impact on pulmonary vessels of strategies aimed at controlling cell senescence in various contexts.
Collapse
Affiliation(s)
- Emmanuelle Born
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| | - Larissa Lipskaia
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| | - Marielle Breau
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| | - Amal Houssaini
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| | - Delphine Beaulieu
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid).,Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, France (D.B., J.-M.F.)
| | - Elisabeth Marcos
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| | - Remi Pierre
- Plate-forme de Recombinaison Homologue, Institut Cochin, INSERM, Paris, France (R.P., M.D.-C.)
| | - Marcio Do Cruzeiro
- Plate-forme de Recombinaison Homologue, Institut Cochin, INSERM, Paris, France (R.P., M.D.-C.)
| | - Marine Lefevre
- Institut Mutualiste Montsouris, Département anatomopathologie, Paris, France (M.L.)
| | - Genevieve Derumeaux
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| | - Dmitry V Bulavin
- Institute for Research on Cancer and Aging, Nice (IRCAN), France (D.V.B.)
| | - Marion Delcroix
- Clinical Department of Respiratory Diseases, University Hospitals and Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven-University of Leuven, Belgium (M.D., R.Q.)
| | - Rozenn Quarck
- Clinical Department of Respiratory Diseases, University Hospitals and Laboratory of Respiratory Diseases & Thoracic Surgery (BREATHE), Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven-University of Leuven, Belgium (M.D., R.Q.)
| | - Virinder Reen
- MRC London Institute of Medical Sciences (LMS), United Kingdom (V.R., J.G.).,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, United Kingdom (V.R., J.G.)
| | - Jesus Gil
- MRC London Institute of Medical Sciences (LMS), United Kingdom (V.R., J.G.).,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, United Kingdom (V.R., J.G.)
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, France (D.B., J.-M.F.)
| | - Jean-Michel Flaman
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM U1052/CNRS 5286, Université de Lyon, Centre Léon Bérard, France (D.B., J.-M.F.)
| | - Serge Adnot
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid).,Institute for Lung Health, Justus Liebig University, Giessen, Germany (S. Adnot).,Medical Research Center, Liaquat University of Medical and Health Sciences, Jamshoro, Pakistan (S. Abid)
| | - Shariq Abid
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, FHU SENEC, AP-HP, Créteil, France; Université Paris-Est Créteil (UPEC), France (E.B., L.L., M.B., A.H., D.P.B., E.M., G.D., S. Adnot, S. Abid)
| |
Collapse
|
27
|
Bloom SI, Islam MT, Lesniewski LA, Donato AJ. Mechanisms and consequences of endothelial cell senescence. Nat Rev Cardiol 2023; 20:38-51. [PMID: 35853997 PMCID: PMC10026597 DOI: 10.1038/s41569-022-00739-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 95.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/15/2022]
Abstract
Endothelial cells are located at the crucial interface between circulating blood and semi-solid tissues and have many important roles in maintaining systemic physiological function. The vascular endothelium is particularly susceptible to pathogenic stimuli that activate tumour suppressor pathways leading to cellular senescence. We now understand that senescent endothelial cells are highly active, secretory and pro-inflammatory, and have an aberrant morphological phenotype. Moreover, endothelial senescence has been identified as an important contributor to various cardiovascular and metabolic diseases. In this Review, we discuss the consequences of endothelial cell exposure to damaging stimuli (haemodynamic forces and circulating and endothelial-derived factors) and the cellular and molecular mechanisms that induce endothelial cell senescence. We also discuss how endothelial cell senescence causes arterial dysfunction and contributes to clinical cardiovascular diseases and metabolic disorders. Finally, we summarize the latest evidence on the effect of eliminating senescent endothelial cells (senolysis) and identify important remaining questions to be addressed in future studies.
Collapse
Affiliation(s)
- Samuel I Bloom
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Md Torikul Islam
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Lisa A Lesniewski
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
- Veterans Affairs Medical Center-Salt Lake City, Geriatric Research Education and Clinical Center, Salt Lake City, UT, USA
| | - Anthony J Donato
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA.
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA.
- Veterans Affairs Medical Center-Salt Lake City, Geriatric Research Education and Clinical Center, Salt Lake City, UT, USA.
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
28
|
Farkas L, Horowitz JC, Mora AL. How a fibroblast ages: a role for bone morphogenetic protein 4 in protecting lung fibroblasts from senescence in pulmonary fibrosis. Eur Respir J 2022; 60:2201702. [PMID: 36522139 PMCID: PMC10210359 DOI: 10.1183/13993003.01702-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Laszlo Farkas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Jeffrey C Horowitz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Ana L Mora
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
29
|
Florentin J, Zhao J, Tai YY, Sun W, Ohayon LL, O'Neil SP, Arunkumar A, Zhang X, Zhu J, Al Aaraj Y, Watson A, Sembrat J, Rojas M, Chan SY, Dutta P. Loss of Amphiregulin drives inflammation and endothelial apoptosis in pulmonary hypertension. Life Sci Alliance 2022; 5:5/11/e202101264. [PMID: 35732465 PMCID: PMC9218345 DOI: 10.26508/lsa.202101264] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022] Open
Abstract
Pulmonary hypertension (PH) is a vascular disease characterized by elevated pulmonary arterial pressure, leading to right ventricular failure and death. Pathogenic features of PH include endothelial apoptosis and vascular inflammation, which drive vascular remodeling and increased pulmonary arterial pressure. Re-analysis of the whole transcriptome sequencing comparing human pulmonary arterial endothelial cells (PAECs) isolated from PH and control patients identified AREG, which encodes Amphiregulin, as a key endothelial survival factor. PAECs from PH patients and mice exhibited down-regulation of AREG and its receptor epidermal growth factor receptor (EGFR). Moreover, the deficiency of AREG and EGFR in ECs in vivo and in vitro heightened inflammatory leukocyte recruitment, cytokine production, and endothelial apoptosis, as well as diminished angiogenesis. Correspondingly, hypoxic mice lacking Egfr in ECs (cdh5 cre/+ Egfr fl/fl) displayed elevated RVSP and pulmonary remodeling. Computational analysis identified NCOA6, PHB2, and RRP1B as putative genes regulating AREG in endothelial cells. The master transcription factor of hypoxia HIF-1⍺ binds to the promoter regions of these genes and up-regulates their expression in hypoxia. Silencing of these genes in cultured PAECs decreased inflammation and apoptosis, and increased angiogenesis in hypoxic conditions. Our pathway analysis and gene silencing experiments revealed that BCL2-associated agonist of cell death (BAD) is a downstream mediator of AREG BAD silencing in ECs lacking AREG mitigated inflammation and apoptosis, and suppressed tube formation. In conclusion, loss of Amphiregulin and its receptor EGFR in PH is a crucial step in the pathogenesis of PH, promoting pulmonary endothelial cell death, influx of inflammatory myeloid cells, and vascular remodeling.
Collapse
Affiliation(s)
- Jonathan Florentin
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jingsi Zhao
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Yi-Yin Tai
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wei Sun
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Lee L Ohayon
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Scott P O'Neil
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Anagha Arunkumar
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Xinyi Zhang
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Jianhui Zhu
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yassmin Al Aaraj
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Annie Watson
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.,Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen Y Chan
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Partha Dutta
- Division of Cardiology, Department of Medicine, Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA .,Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Kyi P, Hendee K, Hunyenyiwa T, Matus K, Mammoto T, Mammoto A. Endothelial senescence mediates hypoxia-induced vascular remodeling by modulating PDGFB expression. Front Med (Lausanne) 2022; 9:908639. [PMID: 36203755 PMCID: PMC9530050 DOI: 10.3389/fmed.2022.908639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/29/2022] [Indexed: 12/03/2022] Open
Abstract
Uncontrolled accumulation of pulmonary artery smooth muscle cells (PASMCs) to the distal pulmonary arterioles (PAs) is one of the major characteristics of pulmonary hypertension (PH). Cellular senescence contributes to aging and lung diseases associated with PH and links to PH progression. However, the mechanism by which cellular senescence controls vascular remodeling in PH is not fully understood. The levels of senescence marker, p16INK4A and senescence-associated β-galactosidase (SA-β-gal) activity are higher in PA endothelial cells (ECs) isolated from idiopathic pulmonary arterial hypertension (IPAH) patients compared to those from healthy individuals. Hypoxia-induced accumulation of α-smooth muscle actin (αSMA)-positive cells to the PAs is attenuated in p16fl/fl-Cdh5(PAC)-CreERT2 (p16iΔEC) mice after tamoxifen induction. We have reported that endothelial TWIST1 mediates hypoxia-induced vascular remodeling by increasing platelet-derived growth factor (PDGFB) expression. Transcriptomic analyses of IPAH patient lungs or hypoxia-induced mouse lung ECs reveal the alteration of senescence-related gene expression and their interaction with TWIST1. Knockdown of p16INK4A attenuates the expression of PDGFB and TWIST1 in IPAH patient PAECs or hypoxia-treated mouse lungs and suppresses accumulation of αSMA–positive cells to the supplemented ECs in the gel implanted on the mouse lungs. Hypoxia-treated mouse lung EC-derived exosomes stimulate DNA synthesis and migration of PASMCs in vitro and in the gel implanted on the mouse lungs, while p16iΔEC mouse lung EC-derived exosomes inhibit the effects. These results suggest that endothelial senescence modulates TWIST1-PDGFB signaling and controls vascular remodeling in PH.
Collapse
Affiliation(s)
- Priscilla Kyi
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kathryn Hendee
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tendai Hunyenyiwa
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kienna Matus
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Akiko Mammoto
| | - Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
- Tadanori Mammoto
| |
Collapse
|
31
|
Kelekçi S, Yıldız AB, Sevinç K, Çimen DU, Önder T. Perspectives on current models of Friedreich’s ataxia. Front Cell Dev Biol 2022; 10:958398. [PMID: 36036008 PMCID: PMC9403045 DOI: 10.3389/fcell.2022.958398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Friedreich’s ataxia (FRDA, OMIM#229300) is the most common hereditary ataxia, resulting from the reduction of frataxin protein levels due to the expansion of GAA repeats in the first intron of the FXN gene. Why the triplet repeat expansion causes a decrease in Frataxin protein levels is not entirely known. Generation of effective FRDA disease models is crucial for answering questions regarding the pathophysiology of this disease. There have been considerable efforts to generate in vitro and in vivo models of FRDA. In this perspective article, we highlight studies conducted using FRDA animal models, patient-derived materials, and particularly induced pluripotent stem cell (iPSC)-derived models. We discuss the current challenges in using FRDA animal models and patient-derived cells. Additionally, we provide a brief overview of how iPSC-based models of FRDA were used to investigate the main pathways involved in disease progression and to screen for potential therapeutic agents for FRDA. The specific focus of this perspective article is to discuss the outlook and the remaining challenges in the context of FRDA iPSC-based models.
Collapse
Affiliation(s)
| | | | | | | | - Tamer Önder
- *Correspondence: Simge Kelekçi, , ; Tamer Önder,
| |
Collapse
|
32
|
Semen KO, Bast A. Senescence in pulmonary arterial hypertension: is there a link? Curr Opin Pulm Med 2022; 28:303-306. [PMID: 35749795 DOI: 10.1097/mcp.0000000000000879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Cellular senescence has been recognized as a promising target in the treatment of many cardiovascular diseases. The pathways involved in the development of senescence share many similarities with pathobiological mechanisms of pulmonary arterial hypertension (PAH). But the potential of senolytics to improve pulmonary hemodynamics and to reduce vascular remodelling in PAH has thus far not been investigated in depth. RECENT FINDINGS PAH does not seem to be a disease of only young people since the mean age of PAH patients is constantly increasing. Changes in expression of senescence biomarkers related to cell cycle arrest, namely upregulation of the tumour suppressor protein p53 and the cell cycle inhibitors p16ink4A an p21cip1 as well as an increase in apoptosis resistance biomarker Bcl2 (B-cell lymphoma 2) and development of senescence-associated phenotype characterized by excessive production of matrix metalloproteinase 2 and interleukin 6 were demonstrated in PAH patients. Initiatives to link the senescence-modulating effect of certain compounds to clinically relevant outcomes in PAH are still limited. SUMMARY Further exploration of the role of senescence in the pathobiology of PAH may point to new relevant treatment strategies. Identification of the cell-specific senescence biomarkers which can be used in vivo, could further promote identification of clinically relevant pathways and design of clinical studies which will help to establish effective therapeutic use of senolytic compounds.
Collapse
Affiliation(s)
| | - Aalt Bast
- Maastricht University Campus Venlo, Venlo
- Department of Pharmacology and Toxicology, FHML, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
33
|
Suraya R, Nagano T, Ryanto GRT, Effendi WI, Hazama D, Katsurada N, Yamamoto M, Tachihara M, Emoto N, Nishimura Y, Kobayashi K. Budesonide/glycopyrronium/formoterol fumarate triple therapy prevents pulmonary hypertension in a COPD mouse model via NFκB inactivation. Respir Res 2022; 23:173. [PMID: 35761394 PMCID: PMC9238100 DOI: 10.1186/s12931-022-02081-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/07/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a health problem that results in death, commonly due to the development of pulmonary hypertension (PH). Here, by utilizing a mouse model of intratracheal elastase-induced emphysema that presents three different phases of COPD, we sought to observe whether budesonide/glycopyrronium/formoterol fumarate (BGF) triple therapy could prevent COPD-PH in addition to ameliorating COPD progression. METHODS We utilized intratracheal elastase-induced emphysema mouse model and performed experiments in three phases illustrating COPD progression: inflammatory (1 day post-elastase), emphysema (3 weeks post-elastase) and PH (4 weeks post-elastase), while treatments of BGF and controls (vehicle, one-drug, and two-drug combinations) were started in prior to elastase instillation (inflammatory phase), at day 7 (emphysema), or at day 14 (PH phase). Phenotype analyses were performed in each phase. In vitro, A549 cells or isolated mouse lung endothelial cells (MLEC) were treated with TNFα with/without BGF treatment to analyze NFκB signaling and cytokine expression changes. RESULTS We observed significant reductions in the proinflammatory phenotype observed in the lungs and bronchoalveolar lavage fluid (BALF) 1 day after elastase administration in mice treated with BGF compared with that in mice administered elastase alone (BALF neutrophil percentage, p = 0.0011 for PBS/Vehicle vs. PBS/Elastase, p = 0.0161 for PBS/Elastase vs. BGF). In contrast, only BGF treatment significantly ameliorated the elastase-induced emphysematous lung structure and desaturation after three weeks of elastase instillation (mean linear intercept, p = 0.0156 for PBS/Vehicle vs. PBS/Elastase, p = 0.0274 for PBS/Elastase vs. BGF). Furthermore, BGF treatment prevented COPD-PH development, as shown by improvements in the hemodynamic and histological phenotypes four weeks after elastase treatment (right ventricular systolic pressure, p = 0.0062 for PBS/Vehicle vs. PBS/Elastase, p = 0.027 for PBS/Elastase vs. BGF). Molecularly, BGF acts by inhibiting NFκB-p65 phosphorylation and subsequently decreasing the mRNA expression of proinflammatory cytokines in both alveolar epithelial and pulmonary endothelial cells. CONCLUSION Our results collectively showed that BGF treatment could prevent PH in addition to ameliorating COPD progression via the inhibition of inflammatory NFκB signaling.
Collapse
Affiliation(s)
- Ratoe Suraya
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Tatsuya Nagano
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan.
| | - Gusty Rizky Teguh Ryanto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada, Kobe, Japan
| | - Wiwin Is Effendi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Daisuke Hazama
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Naoko Katsurada
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Masatsugu Yamamoto
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Motoko Tachihara
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Noriaki Emoto
- Laboratory of Clinical Pharmaceutical Science, Kobe Pharmaceutical University, 4-19-1 Motoyama Kitamachi, Higashinada, Kobe, Japan.,Division of Cardiovascular Medicine, Department of Internal Medicine,, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Yoshihiro Nishimura
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| | - Kazuyuki Kobayashi
- Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki, Chuo, Kobe, Japan
| |
Collapse
|
34
|
Liu L, Wei Y, Giunta S, He Q, Xia S. Potential Role of Cellular Senescence in Pulmonary Arterial Hypertension. Clin Exp Pharmacol Physiol 2022; 49:1042-1049. [PMID: 35748218 DOI: 10.1111/1440-1681.13696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 10/17/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a rare and chronic lung vasculature disease characterized by pulmonary vasculature remodeling, including abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) and dysfunctional endothelial cells (ECs). Remodeling of the pulmonary vasculature occurs from maturity to senescence, and it has become apparent that cellular senescence plays a central role in the pathogenesis of various degenerative vascular diseases and pulmonary pathologies. Cellular senescence represents a state of stable proliferative arrest accompanied by the senescence-associated secretory phenotype (SASP), which entails the copious secretion of proinflammatory signals in the tissue microenvironment. Evidences show that in PAH patients, higher levels of cytokines, chemokines, and inflammatory mediators can be detected and correlate with clinical outcome. Moreover, senescent cells accrue with age in epithelial, endothelial, fibroblastic, and immunological compartments within human lungs, and evidence showed that ECs and PASMCs in lungs from patients with chronic obstructive pulmonary disease were characterized by a higher number of senescent cells. However, there is little evidence uncovering the molecular pulmonary vasculature senescence in PAH. Herein, we review the cellular senescence in pulmonary vascular remodeling, and emphasize its importance in PAH. We further introduce some signaling pathways which might be involved in vasculature senescence and PAH, with the intent to discuss the possibility of the PAH therapy via targeting cellular senescence and reduce PAH progression and mortality.
Collapse
Affiliation(s)
- Lumei Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Yaqin Wei
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, PR China
| | - Sergio Giunta
- Casa di Cura Prof. Nobili-GHC Garofalo Health Care, Bologna, Italy
| | - Qinghu He
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China.,Hunan University of Medicine, Huaihua, PR China
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, PR China
| |
Collapse
|
35
|
Christou H, Khalil RA. Mechanisms of pulmonary vascular dysfunction in pulmonary hypertension and implications for novel therapies. Am J Physiol Heart Circ Physiol 2022; 322:H702-H724. [PMID: 35213243 PMCID: PMC8977136 DOI: 10.1152/ajpheart.00021.2022] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension (PH) is a serious disease characterized by various degrees of pulmonary vasoconstriction and progressive fibroproliferative remodeling and inflammation of the pulmonary arterioles that lead to increased pulmonary vascular resistance, right ventricular hypertrophy, and failure. Pulmonary vascular tone is regulated by a balance between vasoconstrictor and vasodilator mediators, and a shift in this balance to vasoconstriction is an important component of PH pathology, Therefore, the mainstay of current pharmacological therapies centers on pulmonary vasodilation methodologies that either enhance vasodilator mechanisms such as the NO-cGMP and prostacyclin-cAMP pathways and/or inhibit vasoconstrictor mechanisms such as the endothelin-1, cytosolic Ca2+, and Rho-kinase pathways. However, in addition to the increased vascular tone, many patients have a "fixed" component in their disease that involves altered biology of various cells in the pulmonary vascular wall, excessive pulmonary artery remodeling, and perivascular fibrosis and inflammation. Pulmonary arterial smooth muscle cell (PASMC) phenotypic switch from a contractile to a synthetic and proliferative phenotype is an important factor in pulmonary artery remodeling. Although current vasodilator therapies also have some antiproliferative effects on PASMCs, they are not universally successful in halting PH progression and increasing survival. Mild acidification and other novel approaches that aim to reverse the resident pulmonary vascular pathology and structural remodeling and restore a contractile PASMC phenotype could ameliorate vascular remodeling and enhance the responsiveness of PH to vasodilator therapies.
Collapse
Affiliation(s)
- Helen Christou
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raouf A Khalil
- Division of Vascular and Endovascular Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
36
|
Abstract
Pulmonary hypertension (PH) describes heterogeneous population of patients with a mean pulmonary arterial pressure >20 mm Hg. Rarely, PH presents as a primary disorder but is more commonly part of a complex phenotype associated with comorbidities. Regardless of the cause, PH reduces life expectancy and impacts quality of life. The current clinical classification divides PH into 1 of 5 diagnostic groups to assign treatment. There are currently no pharmacological cures for any form of PH. Animal models are essential to help decipher the molecular mechanisms underlying the disease, to assign genotype-phenotype relationships to help identify new therapeutic targets, and for clinical translation to assess the mechanism of action and putative efficacy of new therapies. However, limitations inherent of all animal models of disease limit the ability of any single model to fully recapitulate complex human disease. Within the PH community, we are often critical of animal models due to the perceived low success upon clinical translation of new drugs. In this review, we describe the characteristics, advantages, and disadvantages of existing animal models developed to gain insight into the molecular and pathological mechanisms and test new therapeutics, focusing on adult forms of PH from groups 1 to 3. We also discuss areas of improvement for animal models with approaches combining several hits to better reflect the clinical situation and elevate their translational value.
Collapse
Affiliation(s)
- Olivier Boucherat
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| | - Vineet Agrawal
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allan Lawrie
- Dept of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK & Insigneo institute for in silico medicine, Sheffield, UK
| | - Sebastien Bonnet
- Pulmonary Hypertension Research Group, Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC, Canada
- Department of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
37
|
Abstract
Pulmonary hypertension is an enigmatic, deleterious disease driven by multiple heterogeneous causes with a burgeoning proportion of older patients with complex, chronic comorbidities without adequate treatment options. The underlying endothelial pathophenotypes that direct vasoconstriction and panvascular remodeling remain both controversial and incompletely defined. This review discusses emerging concepts centered on endothelial senescence in pulmonary vascular disease. This principle proposes a more heterogeneous, dynamic pulmonary endothelium in disease; it provides a potentially unifying feature of endothelial dysfunction in pulmonary hypertension irrespective of cause; and it supports a clinically relevant link between aging and pulmonary hypertension like other chronic illnesses. Thus, taking cues from studies on aging and age-related diseases, we present possible opportunities and barriers to diagnostic and therapeutic targeting of senescence in pulmonary hypertension.
Collapse
Affiliation(s)
- Miranda K Culley
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, PA
| |
Collapse
|
38
|
Lees JG, Napierala M, Pébay A, Dottori M, Lim SY. Cellular pathophysiology of Friedreich's ataxia cardiomyopathy. Int J Cardiol 2022; 346:71-78. [PMID: 34798207 DOI: 10.1016/j.ijcard.2021.11.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/01/2021] [Accepted: 11/12/2021] [Indexed: 12/17/2022]
Abstract
Friedreich's ataxia (FRDA) is a hereditary neuromuscular disorder. Cardiomyopathy is the leading cause of premature death in FRDA. FRDA cardiomyopathy is a complex and progressive disease with no cure or treatment to slow its progression. At the cellular level, cardiomyocyte hypertrophy, apoptosis and fibrosis contribute to the cardiac pathology. However, the heart is composed of multiple cell types and several clinical studies have reported the involvement of cardiac non-myocytes such as vascular cells, autonomic neurons, and inflammatory cells in the pathogenesis of FRDA cardiomyopathy. In fact, several of the cardiac pathologies associated with FRDA including cardiomyocyte necrosis, fibrosis, and arrhythmia, could be contributed to by a diseased vasculature and autonomic dysfunction. Here, we review available evidence regarding the current understanding of cellular mechanisms for, and the involvement of, cardiac non-myocytes in the pathogenesis of FRDA cardiomyopathy.
Collapse
Affiliation(s)
- Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Marek Napierala
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, Victoria 3052, Australia; Department of Surgery, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, School of Medicine, Molecular Horizons, University of Wollongong, New South Wales 2522, Australia; Department of Biomedical Engineering, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia; Department of Surgery, The University of Melbourne, Parkville, Victoria 3010, Australia.
| |
Collapse
|
39
|
Kelly NJ, Chan SY. Pulmonary Arterial Hypertension: Emerging Principles of Precision Medicine across Basic Science to Clinical Practice. Rev Cardiovasc Med 2022; 23:378. [PMID: 36875282 PMCID: PMC9980296 DOI: 10.31083/j.rcm2311378] [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] [Indexed: 11/13/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is an enigmatic and deadly vascular disease with no known cure. Recent years have seen rapid advances in our understanding of the molecular underpinnings of PAH, with an expanding knowledge of the molecular, cellular, and systems-level drivers of disease that are being translated into novel therapeutic modalities. Simultaneous advances in clinical technology have led to a growing list of tools with potential application to diagnosis and phenotyping. Guided by fundamental biology, these developments hold the potential to usher in a new era of personalized medicine in PAH with broad implications for patient management and great promise for improved outcomes.
Collapse
Affiliation(s)
- Neil J Kelly
- Center for Pulmonary Vascular Biology and Medicine and Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute; Division of Cardiology; Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine and Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute; Division of Cardiology; Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
40
|
Roger I, Milara J, Belhadj N, Cortijo J. Senescence Alterations in Pulmonary Hypertension. Cells 2021; 10:3456. [PMID: 34943963 PMCID: PMC8700581 DOI: 10.3390/cells10123456] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is the arrest of normal cell division and is commonly associated with aging. The interest in the role of cellular senescence in lung diseases derives from the observation of markers of senescence in chronic obstructive pulmonary disease (COPD), pulmonary fibrosis (IPF), and pulmonary hypertension (PH). Accumulation of senescent cells and the senescence-associated secretory phenotype in the lung of aged patients may lead to mild persistent inflammation, which results in tissue damage. Oxidative stress due to environmental exposures such as cigarette smoke also promotes cellular senescence, together with additional forms of cellular stress such as mitochondrial dysfunction and endoplasmic reticulum stress. Growing recent evidence indicate that senescent cell phenotypes are observed in pulmonary artery smooth muscle cells and endothelial cells of patients with PH, contributing to pulmonary artery remodeling and PH development. In this review, we analyze the role of different senescence cell phenotypes contributing to the pulmonary artery remodeling process in different PH clinical entities. Different molecular pathway activation and cellular functions derived from senescence activation will be analyzed and discussed as promising targets to develop future senotherapies as promising treatments to attenuate pulmonary artery remodeling in PH.
Collapse
Affiliation(s)
- Inés Roger
- Centro de Investigación en Red Enfermedades Respiratorias CIBERES, Health Institute Carlos III, 28029 Valencia, Spain;
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Javier Milara
- Centro de Investigación en Red Enfermedades Respiratorias CIBERES, Health Institute Carlos III, 28029 Valencia, Spain;
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Pharmacy Unit, University General Hospital Consortium of Valencia, 46014 Valencia, Spain
| | - Nada Belhadj
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
| | - Julio Cortijo
- Centro de Investigación en Red Enfermedades Respiratorias CIBERES, Health Institute Carlos III, 28029 Valencia, Spain;
- Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain;
- Research and Teaching Unit, University General Hospital Consortium, 46014 Valencia, Spain
| |
Collapse
|
41
|
Lechartier B, Berrebeh N, Huertas A, Humbert M, Guignabert C, Tu L. Phenotypic Diversity of Vascular Smooth Muscle Cells in Pulmonary Arterial Hypertension: Implications for Therapy. Chest 2021; 161:219-231. [PMID: 34391758 DOI: 10.1016/j.chest.2021.08.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/28/2021] [Accepted: 08/05/2021] [Indexed: 10/20/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive incurable condition that is characterized by extensive remodelling of the pulmonary circulation, leading to severe right heart failure and death. Similar to other vascular contractile cells, pulmonary arterial smooth muscle cells (PA-SMCs) play central roles in physiological and pathological vascular remodelling due to their remarkable ability to dynamically modulate their phenotype to ensure contractile and synthetic functions. The dysfunction and molecular mechanisms underlying their contribution to the various pulmonary vascular lesions associated with PAH have been a major focus of research. The aim of this review is to describe the medial and non-medial origins of contractile cells in the pulmonary vascular wall and present evidence of how they contribute to the onset and progression of PAH. We also highlight specific potential target molecules and discuss future directions that are being explored to widen the therapeutic options for the treatment of PAH.
Collapse
Affiliation(s)
- Benoit Lechartier
- Pulmonary Division, Lausanne University Hospital, Lausanne, Switzerland; Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Nihel Berrebeh
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Alice Huertas
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Marc Humbert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France; AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Ly Tu
- Université Paris-Saclay, School of Medicine, Le Kremlin-Bicêtre, France; INSERM UMR_S 999, Hôpital Marie Lannelongue, Le Plessis-Robinson, France.
| |
Collapse
|
42
|
Lawrie A, Francis SE. Frataxin and endothelial cell senescence in pulmonary hypertension. J Clin Invest 2021; 131:149721. [PMID: 34060487 PMCID: PMC8159686 DOI: 10.1172/jci149721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pulmonary hypertension (PH), increased blood pressure within the lungs, is classified into five diagnostic groups based on etiology, with treatment assigned on this basis. Currently, only Group 1 pulmonary arterial hypertension (PAH) and Group 4 chronic thromboembolic PH (CTEPH) have pharmacological treatments available. The role of the endothelial cell in pulmonary hypertension has long been debated, and in this issue of the JCI, Culley et al. present evidence for the reduction in frataxin expression across multiple groups of PH. Reduced frataxin expression led to endothelial cell senescence and associated with the development of PH. Removal of the senescent cells using the senolytic drug Navitoclax in multiple models of PH effectively treated PH, suggesting a new class of treatments that may work beyond Group 1 and Group 4 PH in patients with evidence of pulmonary vascular endothelial senescence.
Collapse
|