1
|
Zhu X, Pang X, Wang X, Guan X, Tang Y, Wang Z, Zhang L, Zheng X, Li F, Mei J, Ou L, Liu Y, Meng Z, Chen Y, Ma C. Super-Enhancer-Driven LncRNA UNC5B-AS1 Inhibits Inflammatory Phenotypic Transition in Pulmonary Artery Smooth Muscle Cells via Lactylation. Arterioscler Thromb Vasc Biol 2025. [PMID: 40336475 DOI: 10.1161/atvbaha.124.322174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
BACKGROUND The phenotypic transition of pulmonary artery smooth muscle cells (PASMCs) is a central pathological alteration in pulmonary artery remodeling, contributing to pulmonary hypertension. Super-enhancers (SEs), characterized by histone modifications and the binding of coactivators, drive the expression of prominent genes that define cellular identity. However, the specific role of SEs, particularly SE-driven long noncoding RNAs, in hypoxia-induced phenotypic plasticity of PASMCs remains unclear. METHODS In this study, the long noncoding RNA UNC5B antisense RNA 1 (UNC5B-AS1) regulated by SEs was screened in hypoxic PASMCs using RNA sequencing and H3K27ac (histone 3 lysine 27 acetylation) chromatin immunoprecipitation sequencing. Overexpression or knockdown of UNC5B-AS1 in vitro was performed to elucidate its role in pulmonary hypertension pathogenesis. A serotype 5 adenovirus-associated virus carrying a conserved functional fragment of UNC5B-AS1 was used to treat pulmonary hypertension in vivo. RESULTS We identified UNC5B-AS1 as an SE-driven long noncoding RNA transcriptionally activated by the transcription factor FOXP3 (forkhead box protein P3), which regulates phenotypic transition in PASMCs. Notably, we demonstrated that UNC5B-AS1 interacts with key glycolytic enzymes in the cytoplasm and likely serves as a molecular scaffold for LRPPRC (leucine-rich PPR motif-containing protein) and oxidative respiratory chain complex IV in mitochondria. Consequently, the deficiency of UNC5B-AS1 in PASMCs promotes the lactylation of promoter regions within inflammatory genes, including those of IL (interleukin)-1β, IL-6, and TNF-α (tumor necrosis factor-α), under hypoxic conditions, ultimately leading to inflammatory phenotypic transition of PASMCs. CONCLUSIONS Our findings identify SE-driven UNC5B-AS1 as a novel regulatory factor in the hypoxia-induced phenotypic transition of PASMCs and suggest that overexpression of UNC5B-AS1 may represent a promising therapeutic strategy for pulmonary hypertension.
Collapse
Affiliation(s)
- Xiangrui Zhu
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Xiangming Pang
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Xiaoying Wang
- College of Pharmacy, Harbin Medical University (Daqing), PR China. (X.W.)
| | - Xiaoyu Guan
- College of Pharmacy, Harbin Medical University, PR China (X.G.)
| | - Yujing Tang
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Zhaosi Wang
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Lixin Zhang
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Xiaodong Zheng
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Fei Li
- College of Basic Medicine, Harbin Medical University (Daqing), PR China. (F.L.)
| | - Jian Mei
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Langlin Ou
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Yuxiang Liu
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Zitong Meng
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Yingli Chen
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
| | - Cui Ma
- College of Medical Laboratory Science and Technology, Harbin Medical University (Daqing), PR China. (X. Zhu, X.P., Y.T., Z.W., L.Z., X. Zheng, J.M., L.O., Y.L., Z.M., Y.C., C.M.)
- Institute of Cardiovascular Diseases, Xiamen Cardiovascular Hospital, School of Medicine, Xiamen University, Fujian Branch of National Clinical Research Center for Cardiovascular Diseases, PR China (C.M.)
| |
Collapse
|
2
|
Shi S, Chen Q, Yang Y, Li Z, Zheng R, Zhang R, Liu Z, Cheng Y. HnRNPA2B1 promotes cardiac ferroptosis via m6A-dependent stabilization of PFN2 mRNA in myocardial ischemia-reperfusion injury. Free Radic Biol Med 2025; 232:231-243. [PMID: 40010516 DOI: 10.1016/j.freeradbiomed.2025.02.038] [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: 09/30/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 02/28/2025]
Abstract
Myocardial ischemia-reperfusion damage (MIRI) is a clinical problem and lacks proven treatment approaches. As a m6A reader, hnRNPA2B1 controls RNA destiny in the pathophysiology of neurodegenerative and cancerous disorders. Recently, we found that the level of hnRNPA2B1 was elevated in patients with myocardial infarction after percutaneous coronary intervention (PCI), which was positively correlated with cTnI. However, the role of hnRNPA2B1 in MIRI is still unknown. In the present study, we investigated the mechanism underlying MIRI-induced ferroptosis by focusing on a novel function of hnRNPA2B1. Our results showed that HnRNPA2B1 was also significantly increased in cardiomyocytes of MIRI models in vitro and in vivo. Genetically deleting hnRNPA2B1 effectively mitigated myocardial injury and cardiac function during MIRI. Silencing hnRNPA2B1 in cardiomyocytes boosted cell survival and decreased ferroptosis by lowering lipid ROS, MDA, Fe2+, and raising GSH, FTH1 levels, while overexpressing hnRNPA2B1 had the opposite impact. Mechanistic investigations revealed that hnRNPA2B1 recognized and interacted with the m6A site of PFN2 mRNA at "AGACU" to enhance the stability of PFN2 mRNA transcripts. Furthermore, PFN2 knockdown resulted in decreased MDA and Fe2+ levels and an increase in FTH1 expression. Importantly, silencing PFN2 attenuated ferroptosis in cardiomyocytes overexpressing hnRNPA2B1 during OGD/R injury. Collectively, hnRNPA2B1 potentially acts as a therapeutic target of MIRI through regulating caridac ferroptosis mediated by m6A-PFN2/FTH1 pathway.
Collapse
Affiliation(s)
- Shuotao Shi
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Qi Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Yang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Zipei Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ruiyan Zheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Rong Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, Zhuhai, Guangdong, China
| | - Zhongqiu Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, Zhuhai, Guangdong, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau.
| | - Yuanyuan Cheng
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
3
|
Liu Y, Wang R, Jiang T. RNA-binding proteins as a molecular link between COPD and pulmonary hypertension. Int J Med Sci 2025; 22:1979-1991. [PMID: 40225856 PMCID: PMC11983306 DOI: 10.7150/ijms.108587] [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: 12/11/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Pulmonary hypertension (PH) is a vascular disease characterized by remodeling of the pulmonary arteries and right heart failure. Chronic obstructive pulmonary disease (COPD) patients often have PH, which can worsen symptoms and raise morbidity and mortality. There are several reasons for increased pulmonary vascular resistance, pulmonary vascular remodeling, and ultimately the development of PH in COPD. These factors include genetics, inflammation caused by chemicals breathed, and changes in the alveoli seen in COPD and its physiology. Genes involved in mRNA conversion, subcellular localization, splicing, and translation are all finely tuned by RBPs in their post-transcriptional regulation. Erythropoietin regulates cytokines, chemokines, proteins, growth factors, and other pro-inflammatory mediators that change the lung microenvironment. Over the past few years, we have learned more about how RBPs act in PH and COPD. Here, we discuss the existing understanding of RBPs' location in the same pathogenic pathways shared by PH and COPD in order to emphasize their potential relevance as disease determinant/biomarker and, consequently, for possible therapeutic targeting.
Collapse
Affiliation(s)
- Yi Liu
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Ran Wang
- Department of respiratory and critical care medicine, the First Affiliated Hospital of Anhui Medical University, 210 Jixi Road, Hefei, Anhui 230022, China
| | - Tao Jiang
- Department of Respiratory and Critical Care Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| |
Collapse
|
4
|
Tang X, Zeng T, Deng W, Zhao W, Liu Y, Huang Q, Deng Y, Xie W, Huang W. Gut microbe-derived betulinic acid alleviates sepsis-induced acute liver injury by inhibiting macrophage NLRP3 inflammasome in mice. mBio 2025; 16:e0302024. [PMID: 39887250 PMCID: PMC11898617 DOI: 10.1128/mbio.03020-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
Sepsis-induced acute liver injury (SALI) is a prevalent and life-threatening complication associated with sepsis. The gut microbiota plays a crucial role in the maintenance of health and the development of diseases. The impact of physical exercise on gut microbiota modulation has been well-documented. However, the potential impact of gut microbiome on exercise training-induced protection against SALI remains uncertain. Here, we discovered exercise training ameliorated SALI and systemic inflammation in septic mice. Notably, gut microbiota pre-depletion abolished the protective effects of exercise training in SALI mice. Fecal microbiota transplantation treatment revealed that exercise training-associated gut microbiota contributed to the beneficial effect of exercise training on SALI. Exercise training modulated the metabolism of Ligilactobacillus and enriched betulinic acid (BA) levels in mice. Functionally, BA treatment conferred protection against SALI by inhibiting the hepatic inflammatory response in mice. BA bound and inactivated hnRNPA2B1, thus suppressing NLRP3 inflammasome activation in macrophages. Collectively, this study reveals gut microbiota is involved in the protective effects of exercise training against SALI, and gut microbiota-derived BA inhibits the hepatic inflammatory response via the hnRNPA2B1-NLRP3 axis, providing a potential therapeutic strategy for SALI. IMPORTANCE Sepsis is characterized by a dysregulated immune response to an infection that leads to multiple organ dysfunction. The occurrence of acute liver injury is frequently observed during the initial stage of sepsis and is directly linked to mortality in the intensive care unit. The preventive effect of physical exercise on SALI is well recognized, yet the underlying mechanism remains poorly elucidated. Exercise training alters the gut microbiome in mice, increasing the abundance of Ligilactobacillus and promoting the generation of BA. Additionally, BA supplementation can suppress the NLRP3 inflammasome activation in macrophages by directly binding to hnRNPA2B1, thereby mitigating SALI. These results highlight the beneficial role of gut microbiota-derived BA in inhibiting the hepatic inflammatory response, which represents a crucial stride toward implementing microbiome-based therapeutic strategies for the clinical management of sepsis.
Collapse
Affiliation(s)
- Xuheng Tang
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Tairan Zeng
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenyan Deng
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanning Zhao
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanan Liu
- Department of Critical Care Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiyu Deng
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Weidang Xie
- Department of Critical Care Medicine, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Huang
- Department of Critical Care Medicine, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
5
|
Tan J, Cao X, Zheng R, Xie S, Yi J, Wang F, Li X, Song L, Zhang W, Fan J, Qin L, Dai A. Celastrol Ameliorates Hypoxia-Induced Pulmonary Hypertension by Regulation of the PDE5-cGMP-PKG Signaling Pathway. Phytother Res 2025; 39:1549-1564. [PMID: 39887769 DOI: 10.1002/ptr.8446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 12/30/2024] [Accepted: 01/19/2025] [Indexed: 02/01/2025]
Abstract
Pulmonary hypertension (PH) is a severe pulmonary vascular disease characterized by poor clinical outcomes and limited therapeutic options. Celastrol (CEL), a natural product derived from Tripterygium wilfordii Hook F, has shown therapeutic potential in PH models, although its mechanisms are not fully understood. This study aims to investigate the role of CEL in PH and explore its potential underlying mechanisms. This study investigates the role of CEL in PH and explores its underlying mechanisms. We evaluated the effects of CEL in a chronic hypoxia-induced PH rat model and hypoxia-stimulated human pulmonary arterial smooth muscle cells (HPASMCs). Bioinformatics and network pharmacology were employed to identify potential targets and pathways, which were then validated through mechanistic and functional analyses. CEL significantly reduced right ventricular systolic pressure, hypertrophy, fibrosis, and dysfunction in hypoxia-induced PH rats. It also decreased proliferating cell nuclear antigen expression and promoted apoptosis in pulmonary arterioles. Our findings suggest that CEL's therapeutic effects are mediated through the modulation of phosphodiesterase 5 (PDE5) and the activation of the cGMP-PKG signaling pathway. In HPASMCs, CEL treatment mirrored the in vivo results, and PDE5 overexpression negated CEL's antiproliferative, antimigratory, and pro-apoptotic effects. CEL ameliorates pulmonary vascular remodeling and right ventricular dysfunction in PH, potentially through the PDE5-cGMP-PKG signaling pathway. These findings position CEL as a promising candidate for PH therapy.
Collapse
Affiliation(s)
- Junlan Tan
- Department of Gerontology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Xianya Cao
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Runxiu Zheng
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Silin Xie
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Jian Yi
- Department of Gerontology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Feiying Wang
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Xia Li
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Lan Song
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Wen Zhang
- Department of Gerontology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Jianmin Fan
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
| | - Li Qin
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
- Laboratory of Stem Cell Regulation With Chinese Medicine and Its Application, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Aiguo Dai
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, China
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
6
|
Su X, Sun Y, Dai A. New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses. Apoptosis 2025:10.1007/s10495-025-02086-0. [PMID: 39979525 DOI: 10.1007/s10495-025-02086-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xianli Su
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Yinhui Sun
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
7
|
Wang J, Liu J, Yang F, Sun Y, Chen J, Liu J, Sun T, Fan R, Pei F, Luo S, Li J, Luo J. GMRSP encoded by lncRNA H19 regulates metabolic reprogramming and alleviates aortic dissection. Nat Commun 2025; 16:1719. [PMID: 39966416 PMCID: PMC11836370 DOI: 10.1038/s41467-025-57011-5] [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: 02/10/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Metabolic disturbances are hallmarks of vascular smooth muscle cell (VSMC) phenotypic transitions, which play a critical role in the pathogenesis of aortic dissection (AD). In this study, we identify and characterize glucose metabolism regulatory protein (GMRSP), a protein encoded by lncRNA H19. Using VSMC-specific GMRSP induction in knock-in mice, adeno-associated virus-mediated GMRSP overexpression, and exosomal GMRSP delivery, we demonstrate significant improvements in AD and mitochondrial dysfunction. Mechanistically, GMRSP inhibits heterogeneous nuclear ribonucleoprotein (hnRNP) A2B1-mediated alternative splicing of pyruvate kinase M (PKM) pre-mRNA, leading to reduced PKM2 production and glycolysis. This reprogramming preserves the contractile phenotype of VSMCs and prevents their transition to a proliferative state. Importantly, pharmacological activation of PKM2 via TEPP-46 abrogates the protective effects of GMRSP in vivo and in vitro. Clinical relevance is shown by elevated plasma PKM2 levels in AD patients, which correlate with poor prognosis. Collectively, these findings indicate GMRSP as a key regulator of VSMC metabolism and phenotypic stability, highlighting its potential as a therapeutic target for AD.
Collapse
MESH Headings
- Animals
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Aortic Dissection/genetics
- Aortic Dissection/metabolism
- Aortic Dissection/pathology
- Mice
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Pyruvate Kinase/metabolism
- Pyruvate Kinase/genetics
- Thyroid Hormones/metabolism
- Thyroid Hormones/genetics
- Thyroid Hormones/blood
- Myocytes, Smooth Muscle/metabolism
- Thyroid Hormone-Binding Proteins
- Male
- Glycolysis
- Membrane Proteins/metabolism
- Membrane Proteins/genetics
- Carrier Proteins/metabolism
- Carrier Proteins/genetics
- Mice, Inbred C57BL
- Alternative Splicing
- Female
- Mitochondria/metabolism
- Gene Knock-In Techniques
- Metabolic Reprogramming
Collapse
Affiliation(s)
- Jizhong Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Ganzhou Hospital, Guangdong Academy of Medical Sciences, Ganzhou, China
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jitao Liu
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Fan Yang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yinghao Sun
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jiaohua Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Jie Liu
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Tucheng Sun
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ruixin Fan
- Department of Cardiac Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Fang Pei
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Ganzhou Hospital, Guangdong Academy of Medical Sciences, Ganzhou, China
| | - Songyuan Luo
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Jie Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Jianfang Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Ganzhou Hospital, Guangdong Academy of Medical Sciences, Ganzhou, China.
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Guangdong Provincial People's Hospital Nanhai Hospital, foshan, China.
| |
Collapse
|
8
|
Zhang JJ, Ye XR, Liu XS, Zhang HL, Qiao Q. Impact of sodium-glucose cotransporter-2 inhibitors on pulmonary vascular cell function and arterial remodeling. World J Cardiol 2025; 17:101491. [PMID: 39866213 PMCID: PMC11755123 DOI: 10.4330/wjc.v17.i1.101491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/02/2024] [Accepted: 12/17/2024] [Indexed: 01/21/2025] Open
Abstract
Sodium-glucose cotransporter-2 (SGLT-2) inhibitors represent a cutting-edge class of oral antidiabetic therapeutics that operate through selective inhibition of glucose reabsorption in proximal renal tubules, consequently augmenting urinary glucose excretion and attenuating blood glucose levels. Extensive clinical investigations have demonstrated their profound cardiovascular efficacy. Parallel basic science research has elucidated the mechanistic pathways through which diverse SGLT-2 inhibitors beneficially modulate pulmonary vascular cells and arterial remodeling. Specifically, these inhibitors exhibit promising potential in enhancing pulmonary vascular endothelial cell function, suppressing pulmonary smooth muscle cell proliferation and migration, reversing pulmonary arterial remodeling, and maintaining hemodynamic equilibrium. This comprehensive review synthesizes current literature to delineate the mechanisms by which SGLT-2 inhibitors enhance pulmonary vascular cell function and reverse pulmonary remodeling, thereby offering novel therapeutic perspectives for pulmonary vascular diseases.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Chinese Academy Medical Sciences, Fuwai Yunnan Hospital, Kunming 650000, Yunnan Province, China
- Kunming Medical University, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Xue-Rui Ye
- Chinese Academy Medical Sciences, Fuwai Yunnan Hospital, Kunming 650000, Yunnan Province, China
- Kunming Medical University, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China
| | - Xue-Song Liu
- Department of Biochemistry, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Hao-Ling Zhang
- Department of Biomedical Science, Advanced Medical and Dental Institute, University Sains Malaysia, Penang 13200, Malaysia
| | - Qian Qiao
- Chinese Academy Medical Sciences, Fuwai Yunnan Hospital, Kunming 650000, Yunnan Province, China
- Kunming Medical University, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming 650000, Yunnan Province, China.
| |
Collapse
|
9
|
Li L, Xie S, Deng W. RNA binding proteins: Mechanistic considerations and perspectives in controlling cardiovascular diseases. Eur J Pharmacol 2025; 987:177101. [PMID: 39613174 DOI: 10.1016/j.ejphar.2024.177101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) are becoming serious disease that endangering human health due to the increasing morbidity and mortality, and many molecular targets are involved in this complex pathologic process. Recently, RNA-binding proteins (RBPs) have received potential attention as a promising targets for preventing CVDs, including myocardial hypertrophy, dilated cardiomyopathy (DCM), myocardial fibrosis, and pulmonary hypertension (PH). As important regulators of RNA metabolism, RBPs play important roles in all steps of the gene expression cascade,and affect the occurrence and development of various diseases. In this review, we discuss the regulatory role of RBPs on various CVDs at the post transcriptional modification level based on current research. We also highlight the existing and potential RNA-based therapeutics that could impact future CVD treatments.
Collapse
Affiliation(s)
- Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
10
|
Jacob AG, Moutsopoulos I, Petchey A, Kollyfas R, Knight-Schrijver VR, Mohorianu I, Sinha S, Smith CWJ. RNA binding protein with multiple splicing (RBPMS) promotes contractile phenotype splicing in human embryonic stem cell-derived vascular smooth muscle cells. Cardiovasc Res 2024; 120:2104-2116. [PMID: 39248180 PMCID: PMC11646123 DOI: 10.1093/cvr/cvae198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 06/12/2024] [Accepted: 07/14/2024] [Indexed: 09/10/2024] Open
Abstract
AIMS Differentiated vascular smooth muscle cells (VSMCs) express a unique network of mRNA isoforms via smooth muscle-specific alternative pre-mRNA splicing (SM-AS) in functionally critical genes, including those comprising the contractile machinery. We previously described RNA Binding Protein with Multiple Splicing (RBPMS) as a potent driver of differentiated SM-AS in the rat PAC1 VSMC cell line. What is unknown is how RBPMS affects VSMC phenotype and behaviour. Here, we aimed to dissect the role of RBPMS in SM-AS in human cells and determine the impact on VSMC phenotypic properties. METHODS AND RESULTS We used human embryonic stem cell-derived VSMCs (hESC-VSMCs) as our platform. hESC-VSMCs are inherently immature, and we found that they display only partially differentiated SM-AS patterns while RBPMS protein levels are low. We found that RBPMS over-expression induces SM-AS patterns in hESC-VSMCs akin to the contractile tissue VSMC splicing patterns. We present in silico and experimental findings that support RBPMS' splicing activity as mediated through direct binding and via functional cooperativity with splicing factor RBFOX2 on a significant subset of targets. We also demonstrate that RBPMS can alter the motility and the proliferative properties of hESC-VSMCs to mimic a more differentiated state. CONCLUSION Overall, this study emphasizes a critical role for RBPMS in establishing the contractile phenotype splicing programme of human VSMCs.
Collapse
Affiliation(s)
- Aishwarya G Jacob
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | | | - Alex Petchey
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Rafael Kollyfas
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | | | - Irina Mohorianu
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Sanjay Sinha
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | | |
Collapse
|
11
|
Ruffenach G, Medzikovic L, Aryan L, Sun W, Lertpanit L, O'Connor E, Dehghanitafti A, Hatamnejad MR, Li M, Reddy ST, Eghbali M. Intestinal IFNα4 promotes 15-HETE diet-induced pulmonary hypertension. Respir Res 2024; 25:419. [PMID: 39609844 PMCID: PMC11606228 DOI: 10.1186/s12931-024-03046-z] [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: 07/01/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
OBJECTIVES Pulmonary arterial hypertension (PAH) is characterized by the remodeling of the pulmonary vascular bed leading to elevation of the pulmonary arterial pressure. Oxidized fatty acids, such as hydroxyeicosatetraenoic acids (HETEs), play a critical role in PAH. We have previously established that dietary supplementation of 15-HETE is sufficient to cause PH in mice, suggesting a role for the gut-lung axis. However, the mechanisms are not known. APPROACH Analysis of RNA-seq data obtained from the lungs and intestines of mice on 15-HETE diet together with transcriptomic data from PAH patient lungs identified IFN inducible protein 44 (IFI44) as the only gene significantly upregulated in mice and humans. We demonstrate that IFI44 is also significantly increased in PBMCs from PAH patients. In mice, 15-HETE diet enhances IFI44 and its inducer IFN⍺4 expression sequentially in the intestine first and then in the lungs. IFI44 expression in PAH is highly correlated with expression of Tumor Necrosis Factor Related Apoptosis Inducing Ligand (TRAIL), which is upregulated in CD8 cells in PH lungs of both mice and humans. We show that IFNα4 produced by intestinal epithelial cells facilitates IFI44 expression in CD8 cells. Finally, we demonstrate that IFN receptor 1-KO in mice do not develop PH on 15-HETE diet. In addition, silencing IFI44 expression in the lungs of mice on 15-HETE diet prevents the development of PH and is associated with significantly lower expression of IFI44 and TRAIL in CD8 cells in the lungs. CONCLUSION Our data reveal a novel gut-lung axis driven by 15-HETE in PH.
Collapse
Affiliation(s)
- Grégoire Ruffenach
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA.
| | - Lejla Medzikovic
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Laila Aryan
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Wasila Sun
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Long Lertpanit
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-7115, USA
| | - Ellen O'Connor
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-7115, USA
| | - Ateyeh Dehghanitafti
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Mohammad Reza Hatamnejad
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Min Li
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA
| | - Srinivasa T Reddy
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095-7115, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, CHS BH-550 CHS, Los Angeles, CA, 90095-7115, USA.
| |
Collapse
|
12
|
Zhang M, Li L, Zhang W, Li M, Yan G, Tang C. TG2 participates in pulmonary vascular remodelling by regulating the senescence of pulmonary artery smooth muscle cells. Cell Signal 2024; 121:111296. [PMID: 39009200 DOI: 10.1016/j.cellsig.2024.111296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
Pulmonary hypertension (PH) is a severe cardiovascular disease characterised by pulmonary vascular remodelling. The pivotal role of cellular senescence in vascular remodelling has been acknowledged. Transglutaminase type 2 (TG2), a calcium-dependent enzyme, is intricately linked to both cellular senescence and PH. However, the precise mechanisms underlying the involvement of TG2 in PH remain unclear. In this study, we explored the expression of TG2 and the cellular senescence marker p16INK4a in the pulmonary vasculature of mice with PH induced by hypoxia combined with SU5416. Our findings revealed upregulation of both TG2 and p16INK4a expression in the pulmonary vasculature of PH mice. Additionally, a notable increase in TG2 expression was observed in senescent pulmonary artery smooth muscle cells (PASMC). To delve deeper, we employed proteomic sequencing to reveal seven genes associated with cellular senescence, with a subsequent focus on MAPK14. Our investigation revealed that TG2 regulates senescence in PASMC by modulating the phosphorylation levels of MAPK14. Additionally, in the context of hypoxia combined with SU5416, our observations revealed a noteworthy reduction in both pulmonary vascular remodelling and senescent manifestations in smooth muscle-specific TG2 knockout mice compared with their wild-type counterparts. In summary, our findings indicate that TG2 deficiency lowers the senescence levels of PASMC by inhibiting the activity of MAPK14. This inhibition of senescence in the pulmonary vasculature of PH mice helps to decelerate the progression of pulmonary vascular remodelling and consequently hinders the onset and development of PH.
Collapse
Affiliation(s)
- Minhao Zhang
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| | - Linqing Li
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Wenkang Zhang
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Mingkang Li
- School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|
13
|
Wu D, Wang S, Wang F, Zhang Q, Zhang Z, Li X. Lactate dehydrogenase A (LDHA)-mediated lactate generation promotes pulmonary vascular remodeling in pulmonary hypertension. J Transl Med 2024; 22:738. [PMID: 39103838 PMCID: PMC11302077 DOI: 10.1186/s12967-024-05543-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND High levels of lactate are positively associated with prognosis and mortality in pulmonary hypertension (PH). Lactate dehydrogenase A (LDHA) is a key enzyme for the production of lactate. This study is undertaken to investigate the role and molecular mechanisms of lactate and LDHA in PH. METHODS Lactate levels were measured by a lactate assay kit. LDHA expression and localization were detected by western blot and Immunofluorescence. Proliferation and migration were determined by CCK8, western blot, EdU assay and scratch-wound assay. The right heart catheterization and right heart ultrasound were measured to evaluate cardiopulmonary function. RESULTS In vitro, we found that lactate promoted proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) in an LDHA-dependent manner. In vivo, we found that LDHA knockdown reduced lactate overaccumulation in the lungs of mice exposed to hypoxia. Furthermore, LDHA knockdown ameliorated hypoxia-induced vascular remodeling and right ventricular dysfunction. In addition, the activation of Akt signaling by hypoxia was suppressed by LDHA knockdown both in vivo and in vitro. The overexpression of Akt reversed the inhibitory effect of LDHA knockdown on proliferation in PASMCs under hypoxia. Finally, LDHA inhibitor attenuated vascular remodeling and right ventricular dysfunction in Sugen/hypoxia mouse PH model, Monocrotaline (MCT)-induced rat PH model and chronic hypoxia-induced mouse PH model. CONCLUSIONS Thus, LDHA-mediated lactate production promotes pulmonary vascular remodeling in PH by activating Akt signaling pathway, suggesting the potential role of LDHA in regulating the metabolic reprogramming and vascular remodeling in PH.
Collapse
Affiliation(s)
- Daiqian Wu
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, 610014, PR China
| | - Shuo Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, 400042, PR China
| | - Fengxian Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, 400042, PR China
| | - Qing Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, PR China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, 610014, PR China.
| | - Xingbing Li
- Department of Cardiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, PR China.
| |
Collapse
|
14
|
Qiao Y, Mei Y, Xia M, Luo D, Gao L. The role of m6A modification in the risk prediction and Notch1 pathway of Alzheimer's disease. iScience 2024; 27:110235. [PMID: 39040060 PMCID: PMC11261416 DOI: 10.1016/j.isci.2024.110235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 03/17/2024] [Accepted: 05/19/2024] [Indexed: 07/24/2024] Open
Abstract
N6-methyladenosine (m6A) methylation and abnormal immune responses are implicated in neurodegenerative diseases, yet their relationship in Alzheimer's disease (AD) remains unclear. We obtained AD datasets from GEO databases and used AD mouse and cell models, observing abnormal expression of m6A genes in the AD group, alongside disruptions in the immune microenvironment. Key m6A genes (YTHDF2, LRPPRC, and FTO) selected by machine learning were associated with the Notch pathway, with FTO and Notch1 displaying the strongest correlation. Specifically, FTO expression decreased and m6A methylation of Notch1 increased in AD mouse and cell models. We further silenced FTO expression in HT22 cells, resulting in upregulation of the Notch1 signaling pathway. Additionally, increased Notch1 expression in dendritic cells heightened inflammatory cytokine secretion in vitro. These results suggest that reduced FTO expression may contribute to the pathogenesis of AD by activating the Notch1 pathway to interfere with the immune response.
Collapse
Affiliation(s)
- Yingdan Qiao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Yingna Mei
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Minqi Xia
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Deng Luo
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| | - Ling Gao
- Department of Endocrinology & Metabolism, Renmin Hospital of Wuhan University, Wuhan, P.R. China
| |
Collapse
|
15
|
Wang X, Zhi M, Zhao W, Deng J. HNRNPA2B1 promotes oral squamous cell carcinogenesis via m 6A-dependent stabilization of FOXQ1 mRNA stability. IUBMB Life 2024; 76:437-450. [PMID: 38265150 DOI: 10.1002/iub.2808] [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: 11/18/2023] [Accepted: 12/20/2023] [Indexed: 01/25/2024]
Abstract
Oral squamous cell carcinoma (OSCC), as a common type of oral malignancy, has an unclear pathogenesis. N6 methyladenosine (m6A) is a reversible and dynamic process that participates in the modulation of cancer pathogenesis and development. As an m6A recognition protein (reader), heterogeneous nuclear ribonucleoproteins A2/B1 (HNRNPA2B1) show abnormally high expression in cancers. Forkhead box Q1 (FOXQ1), an oncogenic transcription factor, controls multiple biological processes (e.g., embryonic development, cell differentiation, and apoptosis, impacting the initiation and progression of cancers by mediating signaling pathways together with epithelial-mesenchymal transition). Through the Cancer Genome Atlas database screening along with clinical and laboratory experiments, in head and neck squamous cell carcinoma, we found a correlation between HNRNPA2B1 and FOXQ1 gene expression, with shared m6A motifs between HNRNPA2B1 and FOXQ1 mRNA sequences. Silencing or overexpression of HNRNPA2B1 in OSCC cells affected the malignant phenotypes of OSCC cells in vitro, and depletion of HNRNPA2B1 retarded tumor growth in vivo. HNRNPA2B1 could bind to m6A-modified FOXQ1 mRNA to enhance its mRNA stability, resulting in up-regulation of FOXQ1 protein expression. To conclude, HNRNPA2B1 was upregulated in OSCC and enhanced OSCC cell malignant phenotypes by stabilizing m6A-modified FOXQ1 mRNA, eventually aggravating the malignancy and tumorigenicity of OSCC. This study accelerates the recognition of the potency of m6A modification in OSCC and paves the path for OSCC's targeted diagnosis and therapy.
Collapse
Affiliation(s)
- Xi Wang
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, PR China
| | - Min Zhi
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, PR China
| | - Wei Zhao
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, PR China
| | - Jiayin Deng
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, PR China
| |
Collapse
|
16
|
Kong D, Liu J, Lu J, Zeng C, Chen H, Duan Z, Yu K, Zheng X, Zou P, Zhou L, Lv Y, Zeng Q, Lu L, Li J, He Y. HMGB2 Release Promotes Pulmonary Hypertension and Predicts Severity and Mortality of Patients With Pulmonary Arterial Hypertension. Arterioscler Thromb Vasc Biol 2024; 44:e172-e195. [PMID: 38572649 DOI: 10.1161/atvbaha.123.319916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a progressive and life-threatening disease characterized by pulmonary vascular remodeling, which involves aberrant proliferation and apoptosis resistance of the pulmonary arterial smooth muscle cells (PASMCs), resembling the hallmark characteristics of cancer. In cancer, the HMGB2 (high-mobility group box 2) protein promotes the pro-proliferative/antiapoptotic phenotype. However, the function of HMGB2 in PH remains uninvestigated. METHODS Smooth muscle cell (SMC)-specific HMGB2 knockout or HMGB2-OE (HMGB2 overexpression) mice and HMGB2 silenced rats were used to establish hypoxia+Su5416 (HySu)-induced PH mouse and monocrotaline-induced PH rat models, respectively. The effects of HMGB2 and its underlying mechanisms were subsequently elucidated using RNA-sequencing and cellular and molecular biology analyses. Serum HMGB2 levels were measured in the controls and patients with pulmonary arterial (PA) hypertension. RESULTS HMGB2 expression was markedly increased in the PAs of patients with PA hypertension and PH rodent models and was predominantly localized in PASMCs. SMC-specific HMGB2 deficiency or silencing attenuated PH development and pulmonary vascular remodeling in hypoxia+Su5416-induced mice and monocrotaline-treated rats. SMC-specific HMGB2 overexpression aggravated hypoxia+Su5416-induced PH. HMGB2 knockdown inhibited PASMC proliferation in vitro in response to PDGF-BB (platelet-derived growth factor-BB). In contrast, HMGB2 protein stimulation caused the hyperproliferation of PASMCs. In addition, HMGB2 promoted PASMC proliferation and the development of PH by RAGE (receptor for advanced glycation end products)/FAK (focal adhesion kinase)-mediated Hippo/YAP (yes-associated protein) signaling suppression. Serum HMGB2 levels were significantly increased in patients with PA hypertension, and they correlated with disease severity, predicting worse survival. CONCLUSIONS Our findings indicate that targeting HMGB2 might be a novel therapeutic strategy for treating PH. Serum HMGB2 levels could serve as a novel biomarker for diagnosing PA hypertension and determining its prognosis.
Collapse
MESH Headings
- Animals
- HMGB2 Protein/genetics
- HMGB2 Protein/metabolism
- Humans
- Vascular Remodeling
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Pulmonary Artery/metabolism
- Pulmonary Artery/physiopathology
- Pulmonary Artery/pathology
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Rats
- Mice, Inbred C57BL
- Mice
- Cell Proliferation
- Severity of Illness Index
- Signal Transduction
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Arterial Hypertension/physiopathology
- Rats, Sprague-Dawley
- Female
- Cells, Cultured
- Middle Aged
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
Collapse
Affiliation(s)
- Deping Kong
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Jing Liu
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junmi Lu
- Pathology (J. Lu), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Cheng Zeng
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hao Chen
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhenzhen Duan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Ke Yu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Guangdong, China (K.Y.)
| | - Xialei Zheng
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Pu Zou
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liufang Zhou
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Cardiovascular Medicine, The Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi, China (L.Z.)
| | - Yicheng Lv
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Qingye Zeng
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital (D.K., Z.D., Y.L., Q.Z.), Shanghai Jiao Tong University School of Medicine, China
| | - Lin Lu
- Department of Cardiology, Rui Jin Hospital (L.L.), Shanghai Jiao Tong University School of Medicine, China
| | - Jiang Li
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yuhu He
- Departments of Cardiology (D.K., J. Liu, C.Z., H.C., X.Z., P.Z., L.Z., J. Li, Y.H.), The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
17
|
Chen C, Wang C, Liu W, Chen J, Chen L, Luo X, Wu J. Prognostic value and gene regulatory network of CMSS1 in hepatocellular carcinoma. Cancer Biomark 2024; 39:361-370. [PMID: 38160346 PMCID: PMC11191500 DOI: 10.3233/cbm-230209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Cms1 ribosomal small subunit homolog (CMSS1) is an RNA-binding protein that may play an important role in tumorigenesis and development. OBJECTIVE RNA-seq data from the GEPIA database and the UALCAN database were used to analyze the expression of CMSS1 in liver hepatocellular carcinoma (LIHC) and its relationship with the clinicopathological features of the patients. METHODS LinkedOmics was used to identify genes associated with CMSS1 expression and to identify miRNAs and transcription factors significantly associated with CMSS1 by GSEA. RESULTS The expression level of CMSS1 in hepatocellular carcinoma tissues was significantly higher than that in normal tissues. In addition, the expression level of CMSS1 in advanced tumors was significantly higher than that in early tumors. The expression level of CMSS1 was higher in TP53-mutated tumors than in non-TP53-mutated tumors. CMSS1 expression levels were strongly correlated with disease-free survival (DFS) and overall survival (OS) in patients with LIHC, and high CMSS1 expression predicted poorer OS (P< 0.01) and DFS (P< 0.01). Meanwhile, our results suggested that CMSS1 is associated with the composition of the immune microenvironment of LIHC. CONCLUSIONS The present study suggests that CMSS1 is a potential molecular marker for the diagnosis and prognostic of LIHC.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Caiming Wang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
- Department of Operation Room, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Wei Liu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
- Department of Breast, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jiacheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Liang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiangxiang Luo
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jincai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
18
|
Tang L, Cai Q, Wang X, Li X, Li X, Chen L, Yang Y. Canagliflozin ameliorates hypobaric hypoxia-induced pulmonary arterial hypertension by inhibiting pulmonary arterial smooth muscle cell proliferation. Clin Exp Hypertens 2023; 45:2278205. [PMID: 37970663 DOI: 10.1080/10641963.2023.2278205] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a disease with a high mortality and few treatment options to prevent the development of pulmonary vessel remodeling, pulmonary vascular resistance, and right ventricular failure. Canagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, is originally used in diabetes patients which could assist the glucose excretion and decrease blood glucose. Recently, a few studies have reported the protective effect of SGLT2 inhibitor on monocrotaline-induced PAH. However, the effects of canagliflozin on hypobaric hypoxia-induced PAH as well as its mechanism still unclear. In this study, we used hypobaric hypoxia-induced PAH mice model to demonstrate if canagliflozin could alleviate PAH and prevent pulmonary vessel remodeling. We found that daily canagliflozin administration significantly improved survival in mice with hypobaric hypoxia-induced PAH compared to vehicle control. Canagliflozin treatment significantly reduced right ventricular systolic pressure and increased pulmonary acceleration time determined by hemodynamic assessments. Canagliflozin significantly reduced medial wall thickening and decreased muscularization of pulmonary arterioles compared to vehicle treated mice. In addition, canagliflozin inhibited the proliferation and migration of pulmonary arterial smooth muscle cells through suppressing glycolysis and reactivating AMP-activated protein kinase signaling pathway under hypoxia condition. In summary, our findings suggest that canagliflozin is sufficient to inhibit pulmonary arterial remodeling which is a potential therapeutic strategy for PAH treatment.
Collapse
Affiliation(s)
- Luxun Tang
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Qi Cai
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiao Wang
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Xiaoyu Li
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Xiuchuan Li
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, China
| | - Lianglong Chen
- Department of Cardiology, Fujian Medical Center for Cardiovascular Diseases, Fujian Institute of Coronary Heart Disease, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yongjian Yang
- Department of Cardiovascular Medicine, The General Hospital of Western Theater Command PLA, Chengdu, China
| |
Collapse
|
19
|
Qin Y, Yan G, Qiao Y, Wang D, Tang C. Identification of hub genes based on integrated analysis of single-cell and microarray transcriptome in patients with pulmonary arterial hypertension. BMC Genomics 2023; 24:788. [PMID: 38110868 PMCID: PMC10729354 DOI: 10.1186/s12864-023-09892-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a devastating chronic cardiopulmonary disease without an effective therapeutic approach. The underlying molecular mechanism of PAH remains largely unexplored at single-cell resolution. METHODS Single-cell RNA sequencing (scRNA-seq) data from the Gene Expression Omnibus (GEO) database (GSE210248) was included and analyzed comprehensively. Additionally, microarray transcriptome data including 15 lung tissue from PAH patients and 11 normal samples (GSE113439) was also obtained. Seurat R package was applied to process scRNA-seq data. Uniform manifold approximation and projection (UMAP) was utilized for dimensionality reduction and cluster identification, and the SingleR package was performed for cell annotation. FindAllMarkers analysis and ClusterProfiler package were applied to identify differentially expressed genes (DEGs) for each cluster in GSE210248 and GSE113439, respectively. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genome (KEGG) were used for functional enrichment analysis of DEGs. Microenvironment Cell Populations counter (MCP counter) was applied to evaluate the immune cell infiltration. STRING was used to construct a protein-protein interaction (PPI) network of DEGs, followed by hub genes selection through Cytoscape software and Veen Diagram. RESULTS Nineteen thousand five hundred seventy-six cells from 3 donors and 21,896 cells from 3 PAH patients remained for subsequent analysis after filtration. A total of 42 cell clusters were identified through UMAP and annotated by the SingleR package. 10 cell clusters with the top 10 cell amounts were selected for consequent analysis. Compared with the control group, the proportion of adipocytes and fibroblasts was significantly reduced, while CD8+ T cells and macrophages were notably increased in the PAH group. MCP counter revealed decreased distribution of CD8+ T cells, cytotoxic lymphocytes, and NK cells, as well as increased infiltration of monocytic lineage in PAH lung samples. Among 997 DEGs in GSE113439, module 1 with 68 critical genes was screened out through the MCODE plug-in in Cytoscape software. The top 20 DEGs in each cluster of GSE210248 were filtered out by the Cytohubba plug-in using the MCC method. Eventually, WDR43 and GNL2 were found significantly increased in PAH and identified as the hub genes after overlapping these DEGs from GSE210248 and GSE113439. CONCLUSION WDR43 and GNL2 might provide novel insight into revealing the new molecular mechanisms and potential therapeutic targets for PAH.
Collapse
Affiliation(s)
- Yuhan Qin
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Gaoliang Yan
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People's Republic of China.
| | - Yong Qiao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Dong Wang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, People's Republic of China.
| |
Collapse
|
20
|
Ruffenach G, Medzikovic L, Sun W, Hong J, Eghbali M. Functions of RNA-Binding Proteins in Cardiovascular Disease. Cells 2023; 12:2794. [PMID: 38132114 PMCID: PMC10742114 DOI: 10.3390/cells12242794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023] Open
Abstract
Gene expression is under tight regulation from the chromatin structure that regulates gene accessibility by the transcription machinery to protein degradation. At the transcript level, this regulation falls on RNA-binding proteins (RBPs). RBPs are a large and diverse class of proteins involved in all aspects of a transcript's lifecycle: splicing and maturation, localization, stability, and translation. In the past few years, our understanding of the role of RBPs in cardiovascular diseases has expanded. Here, we discuss the general structure and function of RBPs and the latest discoveries of their role in pulmonary and systemic cardiovascular diseases.
Collapse
Affiliation(s)
- Grégoire Ruffenach
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA (W.S.)
| | - Lejla Medzikovic
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA (W.S.)
| | - Wasila Sun
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA (W.S.)
| | - Jason Hong
- Department of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA (W.S.)
| |
Collapse
|
21
|
Aharon-Yariv A, Wang Y, Ahmed A, Delgado-Olguín P. Integrated small RNA, mRNA and protein omics reveal a miRNA network orchestrating metabolic maturation of the developing human heart. BMC Genomics 2023; 24:709. [PMID: 37996818 PMCID: PMC10668469 DOI: 10.1186/s12864-023-09801-8] [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: 08/01/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND As the fetal heart develops, cardiomyocyte proliferation potential decreases while fatty acid oxidative capacity increases in a highly regulated transition known as cardiac maturation. Small noncoding RNAs, such as microRNAs (miRNAs), contribute to the establishment and control of tissue-specific transcriptional programs. However, small RNA expression dynamics and genome-wide miRNA regulatory networks controlling maturation of the human fetal heart remain poorly understood. RESULTS Transcriptome profiling of small RNAs revealed the temporal expression patterns of miRNA, piRNA, circRNA, snoRNA, snRNA and tRNA in the developing human heart between 8 and 19 weeks of gestation. Our analysis demonstrated that miRNAs were the most dynamically expressed small RNA species throughout mid-gestation. Cross-referencing differentially expressed miRNAs and mRNAs predicted 6200 mRNA targets, 2134 of which were upregulated and 4066 downregulated as gestation progressed. Moreover, we found that downregulated targets of upregulated miRNAs, including hsa-let-7b, miR-1-3p, miR-133a-3p, miR-143-3p, miR-499a-5p, and miR-30a-5p predominantly control cell cycle progression. In contrast, upregulated targets of downregulated miRNAs, including hsa-miR-1276, miR-183-5p, miR-1229-3p, miR-615-3p, miR-421, miR-200b-3p and miR-18a-3p, are linked to energy sensing and oxidative metabolism. Furthermore, integrating miRNA and mRNA profiles with proteomes and reporter metabolites revealed that proteins encoded in mRNA targets and their associated metabolites mediate fatty acid oxidation and are enriched as the heart develops. CONCLUSIONS This study presents the first comprehensive analysis of the small RNAome of the maturing human fetal heart. Our findings suggest that coordinated activation and repression of miRNA expression throughout mid-gestation is essential to establish a dynamic miRNA-mRNA-protein network that decreases cardiomyocyte proliferation potential while increasing the oxidative capacity of the maturing human fetal heart. Our results provide novel insights into the molecular control of metabolic maturation of the human fetal heart.
Collapse
Affiliation(s)
- Adar Aharon-Yariv
- Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G0A4, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yaxu Wang
- Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G0A4, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Abdalla Ahmed
- Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G0A4, Canada
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Paul Delgado-Olguín
- Translational Medicine, The Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G0A4, Canada.
- Department of Molecular Genetics, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Heart & Stroke, Richard Lewar Centre of Excellence, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Lu Y, Zou R, Gu Q, Wang X, Zhang J, Ma R, Wang T, Wu J, Feng J, Zhang Y. CRNDE mediated hnRNPA2B1 stability facilitates nuclear export and translation of KRAS in colorectal cancer. Cell Death Dis 2023; 14:611. [PMID: 37716979 PMCID: PMC10505224 DOI: 10.1038/s41419-023-06137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/18/2023]
Abstract
Development of colorectal cancer (CRC) involves activation of Kirsten rat sarcoma viral oncogene homolog (KRAS) signaling. However, the post-transcriptional regulation of KRAS has yet to be fully characterized. Here, we found that the colorectal neoplasia differentially expressed (CRNDE)/heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1) axis was notably elevated in CRC and was strongly associated with poor prognosis of patients, while also significantly promoting CRC cell proliferation and metastasis both in vitro and in vivo. Furthermore, CRNDE maintained the stability of hnRNPA2B1 protein by inhibiting E3 ubiquitin ligase TRIM21 mediated K63 ubiquitination-dependent protein degradation. CRNDE/hnRNPA2B1 axis facilitated the nuclear export and translation of KRAS mRNA, which specifically activated the MAPK signaling pathway, eventually accelerating the malignant progression of CRC. Our findings provided insight into the regulatory network for stable hnRNPA2B1 protein expression, and the molecular mechanisms by which the CRNDE/hnRNPA2B1 axis mediated KRAS nucleocytoplasmic transport and translation, deeply underscoring the bright future of hnRNPA2B1 as a promising biomarker and therapeutic target for CRC. By hindering hnRNPA2B1 from binding to the E3 ubiquitin ligase TRIM21, whose mediated ubiquitin-dependent degradation was thereby inhibited, CRNDE protected the stability of hnRNPA2B1's high protein expression in CRC. Supported by the high level of the oncogenic molecule CRNDE, hnRNPA2B1 bound to KRAS mRNA and promoted KRAS mRNA nucleus export to enter the ribosomal translation program, subsequently activating the MAPK signaling pathway and ultimately accelerating the malignant progression of CRC.
Collapse
Affiliation(s)
- Ya Lu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Renrui Zou
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Quan Gu
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyue Wang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Junying Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Rong Ma
- Research Center of Clinical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Wang
- Department of Cell Biology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Jianzhong Wu
- Research Center of Clinical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jifeng Feng
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuan Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
- Research Center of Clinical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
23
|
Jiang Y, Song S, Liu J, Zhang L, Guo X, Lu J, Li L, Yang C, Fu Q, Zeng B. Epigenetic regulation of programmed cell death in hypoxia-induced pulmonary arterial hypertension. Front Immunol 2023; 14:1206452. [PMID: 37753070 PMCID: PMC10518698 DOI: 10.3389/fimmu.2023.1206452] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/28/2023] [Indexed: 09/28/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe progressive disease that may cause early right ventricular failure and eventual cardiac failure. The pathogenesis of PAH involves endothelial dysfunction, aberrant proliferation of pulmonary artery smooth muscle cells (PASMCs), and vascular fibrosis. Hypoxia has been shown to induce elevated secretion of vascular endothelial growth factor (VEGF), leading to the development of hypoxic PAH. However, the molecular mechanisms underlying hypoxic PAH remain incompletely understood. Programmed cell death (PCD) is a natural cell death and regulated by certain genes. Emerging evidence suggests that apoptotic resistance contributes to the development of PAH. Moreover, several novel types of PCD, such as autophagy, pyroptosis, and ferroptosis, have been reported to be involved in the development of PAH. Additionally, multiple diverse epigenetic mechanisms including RNA methylation, DNA methylation, histone modification, and the non-coding RNA molecule-mediated processes have been strongly linked to the development of PAH. These epigenetic modifications affect the expression of genes, which produce important changes in cellular biological processes, including PCD. Consequently, a better understanding of the PCD processes and epigenetic modification involved in PAH will provide novel, specific therapeutic strategies for diagnosis and treatment. In this review, we aim to discuss recent advances in epigenetic mechanisms and elucidate the role of epigenetic modifications in regulating PCD in hypoxia-induced PAH.
Collapse
Affiliation(s)
- Yuan Jiang
- College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shasha Song
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Jingxin Liu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Liyuan Zhang
- Shanghai Baoxing Biological Equipment Engineering Co., Ltd, Shanghai, China
| | - Xiaofei Guo
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Jiayao Lu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Lie Li
- Shenzhen Reyson Biotechnology Co., Ltd, Shenzhen, China
- Nanjing Evertop Electronics Ltd., Nanjing, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Qiang Fu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Bin Zeng
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| |
Collapse
|
24
|
Gao G, Chen A, Gong J, Lin W, Wu W, Mohammad Ismail Hajary S, Lian G, Luo L, Xie L. Comprehensive analyses of m6A RNA methylation patterns and related immune microenvironment in idiopathic pulmonary arterial hypertension. Front Genet 2023; 14:1222368. [PMID: 37732317 PMCID: PMC10507408 DOI: 10.3389/fgene.2023.1222368] [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: 05/14/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a life-threatening disease with a poor prognosis and high heritability, characterized by elevated pulmonary vascular resistance (PVR) and pulmonary artery pressure. N6-methyladenosine (m6A) RNA modification influences many RNA metabolism pathways. However, the position of m6A methylation regulators in IPAH remains unknown. Therefore, the study aims to disclose the function m6A regulators exert in the pathological mechanisms of IPAH and the immune microenvironment involved. The GSE117261 dataset was downloaded from the Gene Expression Omnibus (GEO) to screen the differentially expressed genes (DEGs) between normal and IPAH samples. Functional and pathway enrichment analyses of DEGs were then conducted by Gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG). We also identified the differentially-expressed m6A (DEm6A) regulators between normal and IPAH samples. Key m6A regulators related to the prediction of IPAH were selected using the random forest model. The results showed that FMR1, RBM15, HNRNPA2B1 and IGFBP3 were upregulated in IPAH. In contrast, LRPPRC was downregulated. The single sample gene set enrichment analysis (ssGSEA) method was then adopted to estimate the immune microenvironment in distinct m6A clusters and m6A phenotype-related genes (PRGs) clusters, respectively. Furthermore, we calculated the m6A score via principal component analysis (PCA), and the Sankey diagram was selected to present the correlation among the m6A clusters, m6A PRGs clusters and m6A score. Finally, quantitative RT-PCR and Western blotting were used to validate the key genes in human pulmonary artery smooth muscle cells (HPASMCs) treated by human platelet-derived growth factor-BB (PDGF-BB). The relative mRNA and protein expression levels of FMR1 were significantly elevated, however, the relative mRNA and protein expression levels of LRPPRC were downregulated. Besides, the relative mRNA level of HNRNPA2B1 was increased. Generally, this bioinformatics analysis might provoke more insights into diagnosing and treating IPAH.
Collapse
Affiliation(s)
- Gufeng Gao
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Ai Chen
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Jin Gong
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Weijun Lin
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Weibin Wu
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Sagor Mohammad Ismail Hajary
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Guili Lian
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Li Luo
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| | - Liangdi Xie
- Department of Geriatrics, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Fujian Hypertension Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Clinical Research Center for Geriatric Hypertension Disease of Fujian Province, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou, China
- Branch of National Clinical Research Center for Aging and Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
- Department of Geriatrics, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fujian, Fuzhou, China
| |
Collapse
|