1
|
Zhang J, Zhang W, Yang Z, Fan B, Wang C, Tian Z. Cinnamaldehyde alleviates pulmonary hypertension by affecting vascular remodeling through the TLR4/NF-kB/HIF-1a pathway. Clin Exp Hypertens 2025; 47:2486829. [PMID: 40171680 DOI: 10.1080/10641963.2025.2486829] [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: 12/17/2024] [Revised: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
OBJECTIVE To investigate the mechanism by which cinnamaldehyde (CA) alleviates pulmonary arterial hypertension (PAH) through the TLR4/NF-kB/HIF-1a pathway. METHODS PAH was induced in rats via SU5416 injection and hypoxia. Hemodynamics (RVMP, RVSP, mPAP) were measured. Histological changes were assessed by HE staining, and protein expressions of α-SMA, Col I, TLR4, p-p65, p65, and HIF-1a were detected by western blot. In vitro, hypoxia-induced HPAECs were treated with CA and TLR4 activator RS09TFA. Cell function was assessed by CCK-8, colony formation, and scratch assays, with VE-Cadherin and α-SMA expression analyzed by western blot. RESULTS PAH rats showed increased RVMP, RVSP, mPAP, and pulmonary artery thickening. CA significantly alleviated lung damage and reduced α-SMA and Col I expression. TLR4/NF-kB/HIF-1a activation with RS09TFA inhibited CA's effects. In vitro, CA mitigated hypoxia-induced HPAEC dysfunction, restoring VE-Cadherin and α-SMA expression, while RS09TFA blocked these effects. CONCLUSION CA alleviates PAH by inhibiting the TLR4/NF-kB/HIF-1a pathway and suppressing vascular remodeling, suggesting its potential as a therapeutic agent for PAH.
Collapse
Affiliation(s)
- Jinbo Zhang
- Department of Preventive Treatment, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Wenxin Zhang
- Department of Culinary and Nutrition, Yantai Vocational College of Culture and Tourism, Yantai, Shandong, China
| | - Zhiyong Yang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Bingbing Fan
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Chunhe Wang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhengkun Tian
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
2
|
Shan L, Tang X, Liu Y, Qi W, Zhao H, Zang L, Li Y, Li X. Targeting BMPER as a therapeutic strategy for pulmonary arterial hypertension. Cell Signal 2025; 133:111880. [PMID: 40383173 DOI: 10.1016/j.cellsig.2025.111880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/19/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Pulmonary arterial hypertension is a life-threatening condition marked by pulmonary vascular remodeling, leading to increased pulmonary artery pressure and right heart hypertrophy. This chronic process involves excessive proliferation and migration of endothelial and smooth muscle cells. Our research, involving both pulmonary arterial hypertension patients and animal models, reveals reduced BMPER levels in cases of pulmonary arterial hypertension. In vitro mechanistic studies are performed using human pulmonary artery endothelial cells and smooth muscle cells. Additionally, we demonstrate BMPER function in vivo through its overexpression via an adeno-associated virus. Our findings indicate that BMPER can attenuate cell proliferation and migration in endothelial cells by inhibiting the PI3K/AKT signaling pathway. Additionally, BMPER reduces smooth muscle cell proliferation and migration by inhibiting BMP4 activity through a paracrine mechanism. Furthermore, BMPER expression is regulated by the transcription factor ERG. Notably, in vivo overexpression of BMPER significantly alleviates the progression of pulmonary arterial hypertension. In summary, our study identifies BMPER as a novel therapeutic target for pulmonary arterial hypertension and provides new insights into the underlying mechanisms of the disease.
Collapse
Affiliation(s)
- Lina Shan
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Xiaofeng Tang
- Department of Respiratory Medicine, Huanggang Central Hospital, Huanggang 439000, China
| | - Yi Liu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua 61700, China
| | - Weiyi Qi
- Department of Respiratory Disease, First Affiliated Hospital of China Medical University, Shen Yang 400042, China
| | - Hainan Zhao
- Department of Nephrology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lie Zang
- Department of Neurology, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Yanwei Li
- Slow Disease Management Center, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Xin Li
- Department of Respiratory Disease, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
3
|
Tang H, Ning K, Wu B, Wang X, He J, Li P, Pan L, Zhang J, He Y, Bian S, Ma X, Zhang J, Liu C, Qin Z, Hu H. Scutellarein ameliorates pulmonary arterial hypertension via sirtuin 1 mediated deacetylation of nicotinamide nucleotide transhydrogenase. Biochem Pharmacol 2025; 237:116932. [PMID: 40189160 DOI: 10.1016/j.bcp.2025.116932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/13/2025]
Abstract
Scutellarein (Sc), a natural flavonoid, holds potential for treating pulmonary arterial hypertension (PAH), yet its mechanisms remain unexplored. This study investigated Sc's therapeutic effects and underlying pathways in PAH. In vivo experiments demonstrated that Sc significantly attenuated right ventricular hypertension, pulmonary arterial remodeling, αSMA expression, and vascular inflammation in PAH models. In vitro, Sc suppressed hypoxia-induced proliferation, migration, inflammation, and pyroptosis in human pulmonary artery smooth muscle cells (HPASMCs). Mechanistically, Sc activated the SIRT1/NAD+ axis to restore mitochondrial homeostasis: it upregulated SIRT1 expression and elevated NAD+ levels by promoting SIRT1-mediated deacetylation of nicotinamide nucleotide transhydrogenase (NNT), thereby enhancing NNT activity. Elevated NAD+ further activated SIRT1, forming a self-reinforcing SIRT1/NNT/NAD+ feedback loop that mitigated hypoxia-induced mitochondrial dysfunction. This study identifies Sc as a novel regulator of the SIRT1-dependent NNT deacetylation pathway, which stabilizes NAD+ homeostasis to counteract HPASMCs dysregulation in PAH. These findings highlight Sc's potential as a therapeutic candidate for PAH, offering insights into targeting mitochondrial-metabolic pathways for vascular remodeling diseases.
Collapse
Affiliation(s)
- Heng Tang
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ke Ning
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Boji Wu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Xuhong Wang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Jingyu He
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Pingping Li
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Lina Pan
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jiawen Zhang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Yi He
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shizhu Bian
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Xingyu Ma
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Jihang Zhang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Chuan Liu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China.
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
4
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Natural products and ferroptosis: A novel approach for heart failure management. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156783. [PMID: 40286752 DOI: 10.1016/j.phymed.2025.156783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/23/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND The discovery of ferroptosis has brought a revolutionary breakthrough in heart failure treatment, and natural products, as a significant source of drug discovery, are gradually demonstrating their extraordinary potential in regulating ferroptosis and alleviating heart failure symptoms. In addition to chemically synthesized small molecule compounds, natural products have attracted attention as an important source for discovering compounds that target ferroptosis in treating heart failure. PURPOSE Systematically summarize and analyze the research progress on improving heart failure through natural products' modulation of the ferroptosis pathway. METHODS By comprehensively searching authoritative databases like PubMed, Web of Science, and China National Knowledge Infrastructure with keywords such as "heart failure", "cardiovascular disease", "heart disease", "ferroptosis", "natural products", "active compounds", "traditional Chinese medicine formulas", "traditional Chinese medicine", and "acupuncture", we aim to systematically review the mechanism of ferroptosis and its link with heart failure. We also want to explore natural small-molecule compounds, traditional Chinese medicine formulas, and acupuncture therapies that can inhibit ferroptosis to improve heart failure. RESULTS In this review, we not only trace the evolution of the concept of ferroptosis and clearly distinguish it from other forms of cell death but also establish a comprehensive theoretical framework encompassing core mechanisms such as iron overload and system xc-/GSH/GPX4 imbalance, along with multiple auxiliary pathways. On this basis, we innovatively link ferroptosis with various types of heart failure, covering classic heart failure types and extending our research to pre-heart failure conditions such as arrhythmia and aortic aneurysm, providing new insights for early intervention in heart failure. Importantly, this article systematically integrates multiple strategies of natural products for interfering with ferroptosis, ranging from monomeric compounds and bioactive components to crude extracts and further to traditional Chinese medicine formulae. In addition, non-pharmacological means such as acupuncture are also included. CONCLUSION This study fills the gap in the systematic description of the relationship between ferroptosis and heart failure and the therapeutic strategies of natural products, aiming to provide patients with more diverse treatment options and promote the development of the heart failure treatment field.
Collapse
Affiliation(s)
- Zeyu Zhang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhihua Yang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shuai Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China
| | - Xianliang Wang
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| | - Jingyuan Mao
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, No.88 Changling Road, Xiqing District, Tianjin 300381, PR China.
| |
Collapse
|
5
|
Wu LW, Chen M, Jiang DJ, Jiang CY, Liu YW, Feng B, Shi CF, Huang X, Zhang X, Xu XH, Zhou XL, Shen Y, Liu TY, Ye LC, He YY, Zhang H, Yan Y. TCF7 enhances pulmonary hypertension by boosting stressed natural killer cells and their interaction with pulmonary arterial smooth muscle cells. Respir Res 2025; 26:202. [PMID: 40442690 PMCID: PMC12121176 DOI: 10.1186/s12931-025-03276-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 05/15/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Pulmonary hypertension (PH) is a life-threatening cardio-pulmonary disorder. Whether natural killer (NK) cells could act as participants in PH and the mechanism by which NK cells moderate pulmonary vascular remodeling has not been fully elucidated. METHODS Single-cell RNA sequencing data from lungs of human pulmonary arterial hypertension (PAH) patients and monocrotaline (MCT)-induced PH rat model were retrieved from GEO database or UCSC Cell Browser. Tcf7 conditional knockout mice and TCF7 overexpression following adeno-associated virus 6 (AAV6) intratracheal delivery in rats were generated. The NK92 cell line and primary human pulmonary artery smooth muscle cells (hPASMCs) were used for in vitro experiments. RESULTS Stressed NK cells were much higher in lungs from human PAH and MCT-induced PH compared to corresponding controls. Of note, TCF7 topped the list differentiating high-stressed from low-stressed human NK cells. TCF7-expressing NK cells displayed higher stress profile than TCF7-deficient cells. Tcf7-deficient NK cells exhibited lower Hsp90aa1 and Hsp90ab1 at transcriptional level and Hsp90 at protein level than Tcf7-expressing cells 24 h post-hypoxia. Mechanistically, TCF7-overexpressing NK cells secrete more SPP1 compared to control NK cells, thus promoting the proliferation and migration of hPASMCs 48 h post-hypoxia. TCF7 overexpression in rats aggravated PH features, while Tcf7 deficiency in mice alleviated pulmonary remodeling possibly due to the manipulation of HSP90 level in NK cells and SPP1 in the microenvironment. CONCLUSIONS TCF7 contributes to the immunopathology of PH possibly through upregulation of stressed NK cells. Under stress conditions, NK cells promote the proliferation and migration of hPASMC through paracrine effects, thereby further promoting vascular remodeling.
Collapse
MESH Headings
- Animals
- Humans
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Rats
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Mice, Knockout
- Male
- Rats, Sprague-Dawley
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/genetics
- Mice, Inbred C57BL
- T Cell Transcription Factor 1/genetics
- T Cell Transcription Factor 1/biosynthesis
- T Cell Transcription Factor 1/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
- Vascular Remodeling
Collapse
Affiliation(s)
- Li-Wei Wu
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Chen
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dai-Ji Jiang
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen-Yu Jiang
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Wei Liu
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Feng
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Xu Huang
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Zhang
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao-He Xu
- School of Pharmacy, Henan University, Kaifeng, China
| | - Xing-Liang Zhou
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Shen
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian-Yu Liu
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lin-Cai Ye
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Hao Zhang
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Yi Yan
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Clinical Research Center for Rare Pediatric Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Research Center for Pediatric Cardiovascular Diseases, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Zhang J, Luo MY, Li NP, Liang N, Yang YH, Zhang YR, Tan GK, Xie T, Gong SX, Wang AP. Histone lactylation-derived prelamin A accelerates pulmonary arterial smooth muscle cells senescence in hypoxia-induced pulmonary hypertension rats. Int Immunopharmacol 2025; 159:114871. [PMID: 40398256 DOI: 10.1016/j.intimp.2025.114871] [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: 02/08/2025] [Revised: 04/14/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
Pulmonary hypertension (PH) is an incurable disease, pulmonary arterial smooth muscle cells (PASMCs) senescence induces PASMCs proliferation by secreting interleukin 6 (IL-6), thereby promoting vascular remodeling. Accumulation of prelamin A is an important initial event at the onset of cellular senescence. Studies have shown that prelamin A has played a crucial role in the senescence-associated cardiovascular disease. However, the role of prelamin A in PH remains to be elucidated. This study aimed to elucidate how prelamin A accelerates PASMCs senescence and its regulatory mechanisms in hypoxia-induced pulmonary hypertension (HPH). Our findings demonstrated increased vascular remodeling and PASMCs senescence in HPH rats, notably, we observed upregulation of prelamin A and histone lactylation in these rats. Inhibiting histone lactylation resulted in decreased prelamin A expression, which consequently mitigated PASMCs senescence and alleviated vascular remodeling in HPH rats. And in 2-DG-treated HPH rats, overexpression of prelamin A reversed defective PASMCs senescence and vascular remodeling. These findings suggest that prelamin A accelerates PASMCs senescence, senescent PASMCs can promote PASMCs proliferation by secreting IL-6, and its upstream regulatory mechanism involves the up-regulation of lactylation modification of histone.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Meng-Yi Luo
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of Laboratory Medicine, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, China
| | - Nan-Ping Li
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Na Liang
- Department of anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Yun-Hua Yang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yu-Rong Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Gang-Kai Tan
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Tong Xie
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of anesthesiology, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421002, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| | - Ai-Ping Wang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
7
|
Deng X, Qiu P, Li X, Hu Y, Que Q, Zhang K, Deng T, Liu Y. Potential of Sivelestat for Pulmonary Arterial Hypertension Treatment: Network Pharmacology-Based Target Identification and Mechanistic Exploration. Drug Des Devel Ther 2025; 19:4123-4138. [PMID: 40416794 PMCID: PMC12103203 DOI: 10.2147/dddt.s507240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 05/04/2025] [Indexed: 05/27/2025] Open
Abstract
Background Sivelestat is a specific neutrophil elastase inhibitor that is currently approved for the treatment of acute lung injury and acute respiratory distress syndrome. Given sivelestat's established anti-inflammatory and antioxidant properties, its efficacy in treating pulmonary arterial hypertension (PAH) remains uncertain. This study aims to investigate the potential of sivelestat as a treatment for PAH. Methods Sivelestat's effects on PAH were evaluated using hypoxia-induced rat models (10% O2, 4 weeks) and pulmonary arterial endothelial/smooth muscle cells (1% O2). Rats received sivelestat (20-100 mg/kg) for 2 weeks, with hemodynamic (RVSP) and vascular remodeling (%WT) assessments. In vitro, sivelestat (50-200 μM) suppressed hypoxia-driven proliferation (CCK-8, EdU), migration (Transwell), and angiogenesis. Molecular validation via qPCR/Western blot confirmed reduced expression of key targets (IGF1R, JAK1, JAK2, PDGFRB). Results Through predictive analysis, we identified 595 potential genes associated with sivelestat in the treatment of PAH. Notably, ERBB2, IGF1R, JAK1, JAK2, PDGFRB, and PTPN11 emerged as key hub genes. In vivo experiments demonstrated that administration of sivelestat at a dose of 100 mg/kg significantly reduced PAH and improved pulmonary vascular remodeling. In vitro experiments indicated that sivelestat effectively decreased the proliferation and migration of PAECs and PASMCs induced by hypoxia. Conclusion Sivelestat has the potential to treat PAH through various targets and pathways. We have initially elucidated the molecular mechanism by which sivelestat acts in the treatment of PAH and have conducted preliminary validation through molecular docking studies and experimental approaches.
Collapse
Affiliation(s)
- Xiaodong Deng
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| | - Pengcheng Qiu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Xin Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, 121000, People’s Republic of China
| | - Yukun Hu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| | - Qing Que
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| | - Kunchi Zhang
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| | - Tianlin Deng
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| | - Yi Liu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, People’s Republic of China
| |
Collapse
|
8
|
Abouzid MR, Kamel I, Twayana AR, Shrestha S, Saleh A, Elshafei S, Jenkins J, Hallak A, Jenkins J. Pulmonary artery denervation in pulmonary hypertension: A comprehensive meta-analysis. Int J Cardiol 2025; 427:133078. [PMID: 40032198 DOI: 10.1016/j.ijcard.2025.133078] [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: 06/16/2024] [Revised: 02/01/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
INTRODUCTION Pulmonary hypertension (PH) is a serious condition characterized by increased pulmonary vascular resistance and elevated pulmonary artery pressure, leading to right heart failure and high mortality rates. Conventional treatments primarily include vasodilators, which provide symptomatic relief but do not effectively reverse the underlying vascular pathology. Pulmonary artery denervation (PADN) has emerged as a novel therapeutic approach targeting the sympathetic nervous system's role in PH. OBJECTIVE This meta-analysis aims to evaluate the impact of PADN on hemodynamic parameters, including mean right atrial pressure (mRAP), mean pulmonary artery pressure (mPAP), and pulmonary vascular resistance (PVR), in patients with pulmonary hypertension. METHODS A comprehensive literature search was conducted across PubMed, Embase, Web of Science, and the Cochrane Library, covering studies published until September 2023. The inclusion criteria focused on studies involving human subjects with PH undergoing PADN, reporting relevant hemodynamic outcomes. Data from 14 studies were analyzed using Review Manager 5.3. Continuous outcomes were pooled using mean differences (MD) with 95 % confidence intervals (CI), and the random effects model was applied where significant heterogeneity was detected. RESULTS The meta-analysis included data from 14 studies comprising 372 patients. PADN resulted in significant reductions in mRAP (MD -1.71 mmHg, 95 % CI -2.34 to -1.08, p < 0.00001, I2 = 15 %), mPAP (MD -10.64 mmHg, 95 % CI -14.33 to -6.95, p < 0.00001, I2 = 84 %), and PVR (MD -4.69 Wood units, 95 % CI -6.55 to -2.83, p < 0.00001, I2 = 93 %). Additionally, there was a significant increase in cardiac output (CO) (MD 0.44 L/min, 95 % CI 0.25 to 0.62, p < 0.00001, I2 = 71 %) and 6-min walk distance (6MWD) (MD 62.4 m, 95 % CI 20.99 to 103.81, p = 0.003, I2 = 94 %). CONCLUSION PADN shows promise as a therapeutic intervention for PH, significantly improving key hemodynamic parameters and exercise capacity. However, the substantial heterogeneity observed among studies highlights the need for standardized procedures and further high-quality, long-term randomized controlled trials to validate these findings and refine patient selection criteria. The results support the potential of PADN to enhance current treatment strategies for pulmonary hypertension.
Collapse
Affiliation(s)
| | - Ibrahim Kamel
- Boston University, Boston, MA, United States of America.
| | - Anu Radha Twayana
- Texas Tech University Health Sciences Center, Odessa, TX, United States of America
| | | | - Amr Saleh
- Yale School of Medicine, New Haven, CT, United States of America
| | - Shorouk Elshafei
- Baptist Hospitals of Southeast Texas, Beaumont, TX, United States of America
| | - Jack Jenkins
- Ochsner Medical Center, New Orleans, LA, United States of America
| | - Ahmad Hallak
- Ochsner Medical Center, New Orleans, LA, United States of America
| | - James Jenkins
- Ochsner Medical Center, New Orleans, LA, United States of America
| |
Collapse
|
9
|
Boehme JT, Datar SA, Sun X, Gong W, Lu Q, Soto J, Smith MA, Garcia-Flores AE, Raff GW, Wang T, Maltepe E, Black SM, Fineman JR. Mechanotransductive stabilization of HIF-1α is inhibited by mitochondrial antioxidant therapy in the setting of pulmonary overcirculation. Sci Rep 2025; 15:16320. [PMID: 40348934 PMCID: PMC12065877 DOI: 10.1038/s41598-025-99062-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 04/15/2025] [Indexed: 05/14/2025] Open
Abstract
In patients with congenital heart disease, the development of pulmonary arterial hypertension (PAH) is based on vascular exposure to abnormal hemodynamic forces. In our work using a large animal model of increased pulmonary blood flow and pressure, we have previously described a pattern of alterations to vascular cell metabolism, mitochondrial function, and mitochondrial redox signaling, paralleling changes in advanced pulmonary vasculopathy states. Based on our findings and emerging literature, we believe that endothelial mitochondria play a central role in integrating and relaying pathologic mechanotransductive signals in abnormal pulmonary hemodynamics. In this manuscript, we demonstrate that exposure of the pulmonary vascular endothelium to aberrant mechanical forces increases production of mitochondrial reactive oxygen species (ROS) and stabilizes the transcription factor Hypoxia Inducible Factor-1α (HIF-1α), and that these changes are associated with impaired endothelial production of Nitric Oxide (NO). We validate that the mitochondrial antioxidant 10-(6'-ubiquinonyl)decyltriphenylphosphonium bromide (MitoQ) can reverse these alterations in vitro, and evaluate the effects of MitoQ treatment in vivo utilizing our large animal shunt model. We find that MitoQ therapy in pulmonary overcirculation decreases the production of mitochondrial ROS, diminishes the mechanically-induced stabilization of HIF-1α, and partially restores vascular reactivity by rescuing endothelial NO production. These findings raise exciting prospects concerning shared pathophysiologic mechanisms and possible common therapeutic targets amongst PAH etiologies.
Collapse
Affiliation(s)
- Jason T Boehme
- Department of Pediatrics, University of California San Francisco, 513 Parnassus Ave., HSE 1401 Box 1346, San Francisco, CA, 94143-2205, USA.
| | - Sanjeev A Datar
- Department of Pediatrics, University of California San Francisco, 513 Parnassus Ave., HSE 1401 Box 1346, San Francisco, CA, 94143-2205, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Wenhui Gong
- Department of Pediatrics, University of California San Francisco, 513 Parnassus Ave., HSE 1401 Box 1346, San Francisco, CA, 94143-2205, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Jamie Soto
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Michael A Smith
- Department of Pediatrics, University of California San Francisco, 513 Parnassus Ave., HSE 1401 Box 1346, San Francisco, CA, 94143-2205, USA
| | - Alejandro E Garcia-Flores
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Gary W Raff
- Department of Surgery, University of California Davis, Davis, CA, 95817, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Emin Maltepe
- Department of Pediatrics, University of California San Francisco, 513 Parnassus Ave., HSE 1401 Box 1346, San Francisco, CA, 94143-2205, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
- Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
- Department of Cellular Biology and Pharmacology, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, 513 Parnassus Ave., HSE 1401 Box 1346, San Francisco, CA, 94143-2205, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| |
Collapse
|
10
|
Yang Y, Zeng Z, Yang Q, Wang H, Zhang H, Yan W, Wang P, Wang C, Su Z, Thangaraju P, Safi SZ, Yang B, Wang Y, Zhou J, Zou Z, Huang Y, Shu S, Xiong C. The Challenge in Burden of Pulmonary Arterial Hypertension: A Perspective From the Global Burden of Disease Study. MedComm (Beijing) 2025; 6:e70175. [PMID: 40276646 PMCID: PMC12019876 DOI: 10.1002/mco2.70175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 02/28/2025] [Accepted: 03/01/2025] [Indexed: 04/26/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) poses significant clinical management challenges due to gaps in understanding its global epidemiology. We analyzed PAH-related disability-adjusted life years (DALYs), deaths, and prevalence from 1990 to 2021. Age-period-cohort models and regression analyses assessed temporal trends and projected burdens to 2050. Globally, PAH-related DALYs declined by 6.6%, but increased by 13.9% in high socio-demographic index (SDI) countries. Middle SDI regions reported the highest DALYs in 1990 and 2021. Deaths rose by 48.5% worldwide, with high SDI nations experiencing a 76.6% surge. Age-standardized rates (ASRs) of DALYs and deaths decreased across SDI countries, with high-middle SDI regions showing the steepest declines. Younger age groups, especially males, had a higher proportion of global DALYs in earlier years, but the burden shifted toward older populations over time, with this trend more pronounced in high-SDI countries. Age-period-cohort analysis revealed declining DALYs in younger ages but rising rates in older cohorts. By 2050, deaths and prevalence are projected to rise, disproportionately affecting females. Significant regional disparities in PAH burden persist, necessitating targeted policies, improved healthcare access, and early detection strategies, especially in underserved areas. Addressing these disparities is critical for mitigating PAH' s global impact.
Collapse
Affiliation(s)
- Yicheng Yang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Center of Respiratory and Pulmonary Vascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of CardiologyAnzhen HospitalBeijingChina
| | - Zhiwei Zeng
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qiaoxi Yang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Huan Wang
- Institute of Child and Adolescent HealthSchool of Public HealthNational Health Commission Key Laboratory of Reproductive Health, Peking UniversityHaidian DistrictBeijingChina
| | - Hanwen Zhang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Center of Respiratory and Pulmonary Vascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wenjie Yan
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Center of Respiratory and Pulmonary Vascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Peizhi Wang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Center for Molecular CardiologyUniversity of ZurichSchlierenZurichSwitzerland
| | - Chuangshi Wang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Medical Research and Biometrics CenterNational Clinical Research Center for Cardiovascular DiseasesFuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Zhanhao Su
- Department of Cardiovascular SurgeryGuangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
| | | | - Sher Zaman Safi
- Faculty of MedicineBioscience & NursingMAHSA UniversitySelangorMalaysia
| | - Beilan Yang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Center of Respiratory and Pulmonary Vascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yaoyao Wang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jingjing Zhou
- Echocardiography Medical CenterBeijing Anzhen Hospital, Capital Medical UniversityBeijingChina
| | - Zhiyong Zou
- Institute of Child and Adolescent HealthSchool of Public HealthNational Health Commission Key Laboratory of Reproductive Health, Peking UniversityHaidian DistrictBeijingChina
| | - Yuan Huang
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Cardiovascular SurgeryFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Songren Shu
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Cardiovascular SurgeryFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Changming Xiong
- State Key Laboratory of Cardiovascular DiseaseFuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Center of Respiratory and Pulmonary Vascular DiseaseFuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
11
|
Lordan J, Weatherald J. Pulmonary arterial hypertension and targeting pulmonary vascular remodelling: are we there yet? Eur Respir J 2025; 65:2500322. [PMID: 40335086 DOI: 10.1183/13993003.00322-2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 03/26/2025] [Indexed: 05/09/2025]
Affiliation(s)
- James Lordan
- Pulmonary Vascular Unit, Freeman Hospital, Newcastle upon Tyne, UK
| | - Jason Weatherald
- Department of Medicine, Division of Pulmonary Medicine, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
12
|
Dong Z, Xue Z, Zhang M, Wang L, Lu Q, Zhao H, Wang X, Nie S, Wei J, Yan H, Fan G, Wang Y, Zhu M. Lingguizhugan decoction inhibit pulmonary hypertension by regulating macrophage IL-6 secretion via IL-33/ST2 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156822. [PMID: 40347929 DOI: 10.1016/j.phymed.2025.156822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Lingguizhugan decoction (LGZG), a typical Chinese herbal formula, has remarkable clinical effects for treating pulmonary hypertension (PH) with unclear ingredients and mechanisms. This study aimed to evaluate the efficacy of LGZG and the mechanism of its active ingredients in treating PH. METHODS The PH patient phenotype was characterized by transcriptomic assay of peripheral blood mononuclear cells (PBMCs) and multiplex cytokine profiling of serum. The active ingredients of LGZG were identified by UPLC-Q-TOF-MS. The monocrotaline (MCT)-induced PH model was performed to evaluate the effects of LGZG and its active ingredients in vivo. The interleukin-33 (IL-33)-induced RAW264.7 cells and the mouse pulmonary artery smooth muscle cells (mPASMCs) were applied to explored phenotype and mechanism of the active ingredients of LGZG by immunofluorescence, western blotting and RT-qPCR in vitro. RESULTS Inflammatory receptor activity in PBMCs and serum interleukin-6 (IL-6) levels were elevated in PH patients. Porinic acid B, cinnamic acid, atractylenolide I and glycyrrhizin (PCAG) were characterized and combined to treat the PH, And the IL-33/Growth stimulation expressed gene 2 (ST2) pathway may be the potential mechanism by transcriptomic analysis of lung tissues. Both in vivo and in vitro experiments confirmed that PCAG inhibited the IL-33/ST2 pathway, reduced macrophage polarization, and suppressed IL-6 secretion. Moreover, ST2 overexpression in macrophages attenuated the inhibitory effect of PCAG. Using a co-culture system of macrophages and mPASMCs, we demonstrated that PCAG reduced mPASMCs phenotypic switching by suppressing macrophage-derived IL-6 secretion. CONCLUSION The active ingredient of LGZG, PCAG, inhibits the progression of PH by preventing macrophage polarization and IL-6 secretion through the IL-33/ST2 pathway.
Collapse
Affiliation(s)
- Zhengwei Dong
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China
| | - Zhifeng Xue
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Zhang
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China
| | - Liming Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Qianqian Lu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China
| | - Huan Zhao
- Henan University of Traditional CM, Zhengzhou 450046, China
| | - Xinlu Wang
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China
| | - Shanshan Nie
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China
| | - Jingjing Wei
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China
| | - Haifeng Yan
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, China.
| | - Yongxia Wang
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China.
| | - Mingjun Zhu
- The First Affiliated Hospital of Henan University of CM, Heart Center/National Regional (Traditional Chinese Medicine) Cardiovascular Diagnosis and Treatment Center, Zhengzhou 450000, China.
| |
Collapse
|
13
|
Lachowicz JA, Steinfort DP, Smallwood NE, Prasad JD. Advances in management of pulmonary fibrosis. Intern Med J 2025. [PMID: 40260907 DOI: 10.1111/imj.70051] [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: 04/30/2024] [Accepted: 03/05/2025] [Indexed: 04/24/2025]
Abstract
Pulmonary fibrosis care, affecting both idiopathic pulmonary fibrosis and other forms of interstitial lung disease (ILD) characterised by fibrosis, has transformed with a range of innovations that affect the diagnosis, treatment and prognosis of this condition. Pharmacotherapeutic options have expanded, with increased indications for the application of effective antifibrotic therapy in non-IPF progressive pulmonary fibrosis as a solo treatment or combined with immunosuppression, emerging evidence for immunomodulatory therapy including biologic agents and greater access to clinical trials. The diagnostic approach to unclassifiable ILD now includes transbronchial lung cryobiopsy, a less invasive method to obtain histopathology with reduced morbidity and mortality compared to surgical lung biopsy. A multidisciplinary approach optimises the care of people with ILD and includes non-pharmacological management, addressing significant comorbidities, symptom care and advanced care planning. This review will summarise recent updates in pulmonary fibrosis management.
Collapse
Affiliation(s)
- Julia A Lachowicz
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Daniel P Steinfort
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Faculty of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Natasha E Smallwood
- Department of Respiratory Medicine, The Alfred, Melbourne, Victoria, Australia
- Faculty of Medicine, Monash University, Melbourne, Victoria, Australia
| | - Jyotika D Prasad
- Department of Respiratory Medicine, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Department of Respiratory Medicine and Lung Transplant Unit, The Alfred, Melbourne, Victoria, Australia
- Honorary Senior Research Fellow, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Li SS, Guo M, Zhao Y, Fan F, Huang S, Yang H, Chen X, Jin X. Intranasal delivery of R8-modified circNFXL1 liposomes ameliorates Su5416-induced pulmonary arterial hypertension in C57BL/6 mice. Respir Res 2025; 26:127. [PMID: 40189516 PMCID: PMC11972480 DOI: 10.1186/s12931-025-03203-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a progressive, life-threatening condition characterized by increased pulmonary vascular resistance and right ventricular hypertrophy (RVH). Current treatments primarily alleviate symptoms but do not effectively target the underlying molecular mechanisms driving the disease. This study aimed to evaluate the therapeutic potential of R8-modified liposomal delivery of circNFXL1, a circular RNA, in a mouse model of PAH. METHODS R8-circNFXL1 liposomes were synthesized and characterized for their physicochemical properties, including encapsulation efficiency. PAH was induced in C57BL/6 mice using a combination of subcutaneous Su5416 administration and hypoxic exposure. Intranasal delivery of R8-circNFXL1 was performed, and therapeutic effects were assessed using echocardiography and hemodynamic measurements. Molecular mechanisms were explored through analysis of the miR-29b/Kcnb1 axis, a regulatory pathway in PAH. RESULTS The R8-circNFXL1 liposomes demonstrated optimal physicochemical properties, including high encapsulation efficiency. Treatment with R8-circNFXL1 significantly reduced RVH, improved cardiac function, and mitigated pulmonary vascular remodeling compared to untreated PAH controls. Molecular analysis revealed that R8-circNFXL1 modulated the miR-29b/Kcnb1 axis, providing insights into its mechanism of action. CONCLUSIONS R8-circNFXL1 liposomes offer a promising, targeted therapeutic strategy for PAH by addressing underlying molecular mechanisms. This approach has potential implications for developing alternative treatments to improve disease management and outcomes in PAH.
Collapse
Affiliation(s)
- Shan-Shan Li
- School of Medicine, Nankai University, Tianjin, China.
| | - Miao Guo
- School of Medicine, Nankai University, Tianjin, China
| | - Ying Zhao
- School of Medicine, Nankai University, Tianjin, China
| | - Feifei Fan
- School of Medicine, Nankai University, Tianjin, China
| | | | | | - Xu Chen
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, China.
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China.
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin, China.
| |
Collapse
|
15
|
Dey T, Zhyvylo I, Jiang L, Olapoju SO, Pena A, Avolio T, Lin D, Goncharov D, Greenland JR, Wolters PJ, DeLisser H, Pullamsetti SS, Kudryashova TV, Goncharova EA. Non-canonical HIPPO-MST1/2 promotes hyper-proliferation of pulmonary vascular cells through CDC20. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.646845. [PMID: 40236147 PMCID: PMC11996459 DOI: 10.1101/2025.04.03.646845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
HIPPO components mammalian Ste20-like protein kinases 1 and 2 (MST1/2) are well described growth suppressors. However, in pulmonary arterial hypertension (PAH), MST1/2 switch their roles and become pro-proliferative and pro-survival molecules, supporting hyper-proliferation of pulmonary artery (PA) smooth muscle cells (PASMCs) and adventitial fibroblasts (PAAFs), remodeling of small PAs, and pulmonary hypertension. Here, we report that MST1/2 promotes hyper-proliferation and apoptosis resistance of human PAH PASMCs and PAAFs by up-regulating cell division cycle protein 20 (CDC20), establishing novel link between HIPPO-MST1/2 and cell cycle regulation in PAH. Authors Contributions conception and design of the work (EAG, SSP, TVK); acquisition, analysis, and interpretation of data (TD, IZ, LJ, SOO, AP, TA, DL, DG, JRG, PJW, HD, TK); drafting and editing the manuscript (EAG, SSP, TVK, JRG, PJW).
Collapse
|
16
|
Song X, Xu Y, Li M, Guan X, Liu H, Zhang J, Sun H, Ma C, Zhang L, Zhao X, Zheng X, Zhu D. SRSF4-Associated ca-circFOXP1 Regulates Hypoxia-Induced PASMC Proliferation by the Formation of R Loop With Host Gene. Arterioscler Thromb Vasc Biol 2025; 45:e118-e135. [PMID: 39973750 DOI: 10.1161/atvbaha.124.322026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a rare and fatal disease, the pathological changes of which include pulmonary arterial smooth muscle cell (PASMC) proliferation, which is the pathological basis of pulmonary vascular remodeling. Studies have demonstrated that chromatin-associated circRNA can regulate a variety of biological processes. However, the role of chromatin-associated circRNA in the proliferation of PH remains largely unexplored. In this study, we aimed to identify the function and mechanism of chromatin-associated circRNA in PASMC proliferation in PH. METHODS The role of chromatin-associated circFOXP1 (ca-circFOXP1) was investigated in hypoxic mouse PASMCs and SuHX (Sugen5416+hypoxia) model mice through the use of antisense oligonucleotide knockdown and adeno-associated virus-mediated knockdown. Through bioinformatic sequence alignment, chromatin isolation by RNA purification, Cell Counting Kit 8, 5-ethynyl-2-deoxyuridine, Western blot, and other experiments, the function and mechanism of ca-circFOXP1 were verified. RESULTS The expression of ca-circFOXP1 was found to be significantly increased in SuHX model mice and hypoxic mouse PASMCs. Moreover, ca-circFOXP1 was found to regulate the level of the host protein FOXP1 (forkhead box protein 1) through the R loop, thereby influencing the phosphorylation activity of SMAD2 (SMAD family member 2) and, consequently, the proliferation of mouse PASMCs. It is noteworthy that the m6A modification was found to promote the formation of the R loop between ca-circFOXP1 and the host gene FOXP1, thereby regulating the expression of the host protein. Furthermore, we have identified that the splicing factor SRSF4 (serine/arginine rich splicing factor 4) can upregulate the expression of ca-circFOXP1 by splicing exons 6 and 9 of FOXP1 pre-mRNA. CONCLUSIONS The results demonstrated that the splicing factor SRSF4 upregulated the expression of ca-circFOXP1, and m6A methylation promoted R-loop formation between ca-circFOXP1 and host genes, regulated the level of host protein FOXP1, and then affected the phosphorylation activity of SMAD2, mediating PASMC proliferation, leading to pulmonary vascular remodeling. These results provide a theoretical basis for further study of the pathological mechanisms of hypoxic PH and may provide certain insights.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Disease Models, Animal
- Pulmonary Artery/pathology
- Pulmonary Artery/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Mice
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Serine-Arginine Splicing Factors/genetics
- Serine-Arginine Splicing Factors/metabolism
- Cells, Cultured
- Signal Transduction
- Smad2 Protein/metabolism
- Male
- Vascular Remodeling
- Mice, Inbred C57BL
- Hypoxia/metabolism
- Hypoxia/genetics
- Phosphorylation
- Cell Hypoxia
- Repressor Proteins
Collapse
Affiliation(s)
- Xinyue Song
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Ya Xu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Mengnan Li
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Xiaoyu Guan
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Huiyu Liu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Jingya Zhang
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Hanliang Sun
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| | - Cui Ma
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Lixin Zhang
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Xijuan Zhao
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- College of Medical Laboratory Science and Technology (C.M., L.Z., X. Zhao), Harbin Medical University (Daqing), P.R. China
| | - Xiaodong Zheng
- Department of Genetic and Cell Biology (X. Zheng), Harbin Medical University (Daqing), P.R. China
| | - Daling Zhu
- College of Pharmacy (X.S., Y.X., M.L., X.G., H.L., H.S., C.M., L.Z., X. Zhao, D.Z.), Harbin Medical University, P.R. China
- Key Laboratory of Cardiovascular Medicine Research, Ministry of Education (D.Z.), Harbin Medical University, P.R. China
- Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.S., Y.X., M.L., X.G., H.L., J.Z., H.S., D.Z.)
| |
Collapse
|
17
|
Xiao Y, Ma R, Liu C, Wei H, Zhou B, Gui C, Yang B, Cao Y, Zhang K, Luo B. Insights into Pulmonary Heart Disease at High Altitude: Hypoxia and PM 2.5 Interactions. Am J Respir Crit Care Med 2025; 211:660-663. [PMID: 39836437 DOI: 10.1164/rccm.202409-1802rl] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Affiliation(s)
- Ya Xiao
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Runxue Ma
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Ce Liu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Huanhuan Wei
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Baofeng Zhou
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Chunyan Gui
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Bo Yang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| | - Yunshan Cao
- Heart, Lung and Vessels Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; and
| | - Kai Zhang
- Department of Environmental Health Sciences, School of Public Health, University at Albany, State University of New York, Rensselaer, New York
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
18
|
Fiorentù G, Bernardinello N, Giulianelli G, Cocconcelli E, Balestro E, Spagnolo P. Pulmonary Hypertension Associated with Interstitial Lung Disease (PH-ILD): Back to the Future. Adv Ther 2025; 42:1627-1641. [PMID: 39969780 PMCID: PMC11929637 DOI: 10.1007/s12325-025-03129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Pulmonary hypertension (PH) is a progressive syndrome characterized by increased pulmonary artery pressure. PH often complicates chronic lung diseases, thus contributing to a substantial disease burden and poor prognosis. The WHO Group 3 Pulmonary Hypertension has many subcategories, including sleep-hypoventilation PH, high altitude-PH, chronic obstructive pulmonary disease (COPD)-PH, and interstitial lung disease (PH-ILD), the latter carrying the worst prognosis. ILD is a heterogeneous group of disorders characterized by cough and shortness of breath and, in progressive forms, irreversible loss of function and respiratory failure. The development of PH in patients with ILD worsens exercise capacity and exertional dyspnea and impairs quality of life. Thus, suspicion and early detection of PH following thorough cardiologic evaluation (i.e., echocardiography, pro-BNP, and right heart catheterization) is paramount for appropriate patient management. For PH secondary to chronic respiratory diseases, current guidelines recommend optimizing the treatment of the underlying respiratory condition and offering long-term oxygen therapy. In recent years, several clinical trials have failed to identify drugs beneficial for group 3 PH. Conversely, the INCREASE trial of inhaled treprostinil has recently provided hope for treating PH-ILD. In this review, we summarize and critically discuss the present and future of the pharmacological management of PH-ILD.
Collapse
Affiliation(s)
- Giordano Fiorentù
- Departement of Cardiac, Thoracic, Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Nicol Bernardinello
- Departement of Cardiac, Thoracic, Vascular Science and Public Health, University of Padova, Padua, Italy.
| | - Giacomo Giulianelli
- Departement of Cardiac, Thoracic, Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Elisabetta Cocconcelli
- Departement of Cardiac, Thoracic, Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Elisabetta Balestro
- Departement of Cardiac, Thoracic, Vascular Science and Public Health, University of Padova, Padua, Italy
| | - Paolo Spagnolo
- Departement of Cardiac, Thoracic, Vascular Science and Public Health, University of Padova, Padua, Italy
| |
Collapse
|
19
|
Ye Y, Zhou M, Yin D, Wang Y, Lin J, Sun J, Wang X, Guo Q. Association of interferon regulator factor 1 upregulation with pulmonary arterial hypertension. J Thorac Dis 2025; 17:1698-1710. [PMID: 40223959 PMCID: PMC11986763 DOI: 10.21037/jtd-2025-390] [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: 02/24/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025]
Abstract
Background Pulmonary arterial hypertension (PAH) is a complex disease that is associated with a poor prognosis. Its pathogenesis is attributed to the inflammatory immune response. Interferon regulator factor 1 (IRF1) is a key downstream regulator of inflammation and cell death. Evidence suggests that IRF1 can promote the proliferation of smooth muscle cells and inhibit lung endothelial regeneration. However, proof for this relationship is lacking, and the exact nature of the potential mechanism underlying the link between IRF1 and PAH remains largely unknown. We aimed to find out whether IRF1 is associated with the progression of PAH. Methods The GSE144274 and GSE243193 datasets were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between PAH and healthy samples were identified and analyzed. Enrichment analysis was performed, and a protein-protein interaction (PPI) network was constructed to identify the hub genes. The relative protein and gene levels of IRF1 were then validated in PAH animal models. Results A total of 271 DEGs were identified from the two data sets. ACTA2, HLA-DRA, HLA-A, PECAM1, HLA-C, IRF1, and CD74 were identified as the hub genes. In our subsequent experiments, we found that IRF1 was upregulated in both PAH rat and mouse models. Conclusions Our findings suggest that IRF1 might be associated with pulmonary hypertension in lung tissue and may thus serve as a therapeutic target in PAH.
Collapse
Affiliation(s)
- Yuheng Ye
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Zhou
- Department of Critical Care Medicine, Hai’an People’s Hospital, Nantong, China
| | - Di Yin
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Yi Wang
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiancheng Lin
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiayan Sun
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaowan Wang
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| | - Qiang Guo
- Department of Critical Care Medicine, The Fourth Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
20
|
Liu C, Meng F. Identification of potential causal genes and drug targets in pulmonary hypertension based on transcriptomic analysis and Mendelian randomization. Postgrad Med J 2025:qgaf044. [PMID: 40156905 DOI: 10.1093/postmj/qgaf044] [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: 11/22/2024] [Revised: 02/16/2025] [Accepted: 03/08/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE Currently, there is no definitive treatment for pulmonary hypertension (PH). This study aims to utilize the GEO database and conduct Mendelian randomization (MR) analysis to identify new genetic targets for PH and investigate their potential pathogenic pathways and therapeutic drugs. METHODS We identified key genes by combining the findings from MR and bioinformatics analyses of GEO datasets. We performed enrichment analysis to explore the functional roles of these key genes. Then, we constructed protein-protein interaction (PPI) and miRNA-mRNA networks to identify interacting proteins and miRNAs. Drug prediction analysis was conducted to propose potential therapeutic drugs. Finally, we validated the results through the GEO dataset, RT-PCR, and western blot experiments. RESULTS The joint analysis utilizing GEO databases and MR analysis identified two key genes, ITGA2B and TSPAN9 that exhibited significance across both analytical methods. The enrichment analysis indicated that the key genes were involved in critical biological functions and pathways, including cell adhesion, platelet activation, and the PI3K-Akt signaling pathway. The PPI and miRNA-mRNA networks further highlighted the significance of the key genes in PH. Drug prediction analysis revealed the potential of the key genes as therapeutic targets. The RT-PCR and western blot experiments validated the above findings. CONCLUSION By integrating bioinformatics and MR analysis, we found that ITGA2B and TSPAN9 have a causal relationship with PH. Our findings offer new insights into the molecular mechanism and potential treatment targets of PH, establishing a basis for future research and clinical applications.
Collapse
Affiliation(s)
- Chengliang Liu
- Department of Respiratory and Critical Care Medicine, Chaohu Hospital of Anhui Medical University, No. 64 Chaohu North Road, Chaohu 238000, China
| | - Fanliang Meng
- Department of Respiratory and Critical Care Medicine, Chaohu Hospital of Anhui Medical University, No. 64 Chaohu North Road, Chaohu 238000, China
| |
Collapse
|
21
|
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
|
22
|
Tan W, Wang Y, Li M, Zhao C, Hu Y, Gao R, Chen Z, Hu L, Li Q. A novel pyridine-2-one AMPK inhibitor: Discovery, mechanism, and in vivo evaluation in a hypoxic pulmonary arterial hypertension rat model. Eur J Med Chem 2025; 286:117266. [PMID: 39826489 DOI: 10.1016/j.ejmech.2025.117266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
AMP-activated protein kinase (AMPK), a heterotrimeric serine-threonine kinase, has been identified as a promising target for regulating vascular remodeling in pulmonary arterial hypertension (PAH) due to its capacity to promote proliferation, autophagy, and anti-apoptosis in pulmonary artery smooth muscle cells (PASMCs). However, research into AMPK inhibitors is very limited. Herein, a virtual screening strategy was employed to identify CHEMBL3780091 as a lead compound for a series of novel AMPK inhibitors by exploring the structure-activity relationship around a specific pyridine-2-one scaffold. Subsequently, the most promising 13a was observed to exhibit excellent AMPK inhibitory activity and favorable anti-proliferative activity against PASMCs through the inhibition of the AMPK signaling pathway in vitro. Moreover, compound 13a significantly reduced right ventricular systolic pressure, attenuated vascular remodeling, and improved right heart function in hypoxia-induced PAH rats in vivo. In conclusion, this study provides a novel and potential lead compound for the study of AMPK inhibitors and a new direction for the development of PAH drugs that focus on improving vascular remodeling.
Collapse
Affiliation(s)
- Wenhua Tan
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yu Wang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Mengqi Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Congke Zhao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Yuanbo Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Ruizhe Gao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China.
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha, 410013, Hunan, China.
| |
Collapse
|
23
|
Chen Z, Cheang I, Zhu X, Shi J, Chen S, Fu Y, Liao S, Gao R, Zhou Y, Li X. Validation of a non-invasive filling pressure (NIFP) device for measuring cardiac pressure and assessing congestion levels in patients with heart failure. Int J Cardiol 2025; 422:132973. [PMID: 39788351 DOI: 10.1016/j.ijcard.2025.132973] [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: 11/13/2024] [Revised: 12/24/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
BACKGROUND Heart failure (HF) is a high-burden clinical syndrome characterized by intravascular and extravascular congestion, impacting patients' outcomes. Current diagnostic methods for assessing intravascular congestion, including right heart catheterization (RHC), have some limitations. There is a need for accurate, stable, and widely applicable non-invasive measurement methods to improve HF diagnosis and treatment. METHODS We conducted non-invasive filling pressure (NIFP) measurements in 74 patients before or after RHC. The correlation between recorded factors and pulmonary arterial wedge pressure (PAWP) values was examined. We tested NIFP's performance as a predictive tool for PAWP using different thresholds. Receiver operating characteristic (ROC) curve analysis and Pearson correlation coefficient were used for data analysis. RESULTS NIFP measurement served as an independent impact factor and showed a definite relationship with PAWP in both univariate and multivariate regression analyses (all p < 0.05). NIFP demonstrated moderate accuracy in predicting PAWP values (all AUC > 0.75), particularly among patients without arrhythmia [AUC for Model 1 (PAWP >15): 0.80; AUC for Model 2 (PAWP >18): 0.85]. CONCLUSIONS The NIFP device represents a promising innovation, offering non-invasive and user-friendly solution for precisely measuring pulmonary arterial wedge pressure (PAWP) and assessing the degree of congestion.
Collapse
Affiliation(s)
- Ziqi Chen
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Iokfai Cheang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xu Zhu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jinjing Shi
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Sitong Chen
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yiyang Fu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Shengen Liao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Rongrong Gao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yanli Zhou
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xinli Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China.
| |
Collapse
|
24
|
Deng X, Que Q, Zhang K, Li B, Yang N, Hu Q, Lv S, Liu Y. Mechanistic insights into the role of EGLN3 in pulmonary vascular remodeling and endothelial dysfunction. Respir Res 2025; 26:61. [PMID: 39985019 PMCID: PMC11844033 DOI: 10.1186/s12931-025-03144-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/09/2025] [Indexed: 02/23/2025] Open
Abstract
Endothelial dysfunction is a pivotal initiating factor in vascular remodeling in pulmonary hypertension. EGLN3, a hypoxia response factor, plays a significant role in cell proliferation and angiogenesis, which are closely related to the pathophysiological conditions of pulmonary hypertension. This study investigates the potential involvement of EGLN3 in the injury response of pulmonary vascular endothelial cells and its contribution to the development of pulmonary arterial hypertension. Research has demonstrated that in patients with pulmonary arterial hypertension and various animal models of the condition, EGLN3 expression is upregulated in the remodeled pulmonary artery endothelium. Notably, the endothelial cell-specific knockout of EGLN3 can decelerate the progression of pulmonary arterial hypertension, whereas its overexpression has the opposite effect. Mechanistic analyses reveal that under hypoxic conditions, JUN initiates the transcription of EGLN3 by binding to its promoter region. Subsequently, EGLN3 interacts with HUR to enhance the stability of EGFR mRNA, thereby activating the PI3K/AKT and MAPK signaling pathways, which ultimately results in endothelial cell damage, proliferation, and migration. These findings suggest that EGLN3 is a critical gene for maintaining endothelial function and vascular homeostasis and holds promise as a novel therapeutic target for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Xiaodong Deng
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Qing Que
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Kunchi Zhang
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Bo Li
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Nianlong Yang
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Qiang Hu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Sheng Lv
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China
| | - Yi Liu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua, 61700, China.
| |
Collapse
|
25
|
Liang Y, Ornatowski W, Lu Q, Sun X, Yegambaram M, Feng A, Dong Y, Aggarwal S, Unwalla HJ, Fineman JR, Black SM, Wang T. Chloroquine Restores eNOS Signaling in Shunt Endothelial Cells via Inhibiting eNOS Uncoupling. Int J Mol Sci 2025; 26:1352. [PMID: 39941119 PMCID: PMC11818845 DOI: 10.3390/ijms26031352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by increased lung vascular stiffness and impaired vessel relaxation, primarily due to reduced nitric oxide (NO) production in endothelial cells. Recent studies indicate that chloroquine, an autophagy inhibitor, may help lower pulmonary arterial pressure and enhance lung vascular function. This study investigates the mechanisms underlying the chloroquine-mediated restoration of NO bioavailability in endothelial cells derived from aortopulmonary shunt lambs, a relevant model for congenital heart defect (CHD)-associated PAH. We found that NO production was significantly reduced in shunt pulmonary artery endothelial cells (PAECs), attributable to decreased levels of tetrahydrobiopterin (BH4) and diminished expression of GTP cyclohydrolase 1 (GCH1), despite a slight increase in endothelial nitric oxide synthase (eNOS) levels. Chloroquine robustly restored endothelial NO production, which correlated with increased BH4 levels and restored GCH1 expression. The mechanistically upregulated carboxyl terminus of Hsp70-interacting protein (CHIP) in shunt PAECs is responsible for heightened GCH1 degradation, and chloroquine disrupted the assembly of the GCH1-HSP70-CHIP complex to preserve cellular GCH1. Similarly, another autophagy inhibitor, bafilomycin A1, demonstrated comparable effects. These findings suggest that autophagy inhibition can effectively enhance NO synthesis in endothelial cells experiencing depleted NO bioavailability, presenting a potential therapeutic strategy for managing PAH.
Collapse
Affiliation(s)
- Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Wojciech Ornatowski
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Yishu Dong
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Hoshang J. Unwalla
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular and Molecular Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
26
|
Deng X, You Y, Lv S, Liu Y. MMP8-mediated vascular remodeling in pulmonary hypertension. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167582. [PMID: 39581558 DOI: 10.1016/j.bbadis.2024.167582] [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: 09/13/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a vascular remodeling disease that impacts the cardiopulmonary system. Due to the currently limited understanding of vascular remodeling, a cure for PAH remains elusive. This study highlights the critical role of the STAT1 (signal transducer and activator of transcription 1)/MMP8 (matrix metallopeptidase 8)/DRP1 (dynamin-related protein 1) axis in vascular remodeling and the pathogenesis of pulmonary hypertension. Notably, MMP8 is significantly elevated in pulmonary arterial endothelial cells and its levels correlate with the severity of the disease. MMP8 binds to and activates DRP1, inducing mitochondrial fragmentation and promoting a malignant phenotype of endothelial cells under hypoxic conditions. Moreover, MMP8 is tightly regulated by STAT1. The knockout of MMP8 attenuates chronic pulmonary vascular remodeling, and drugs targeting MMP8 alleviate pulmonary hypertension and enhance cardiac function. This study offers fresh insights into hypoxia-induced vascular remodeling, laying a theoretical foundation for countering vascular remodeling by directly regulating the STAT1/MMP8/DRP1 axis.
Collapse
Affiliation(s)
- Xiaodong Deng
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua 61700, China
| | - Yong You
- Department of Respiratory department, Huanggang Central Hospital, Huanggang 438000, China
| | - Sheng Lv
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua 61700, China
| | - Yi Liu
- Department of Critical Care Medicine, Panzhihua Central Hospital, Panzhihua 61700, China.
| |
Collapse
|
27
|
Hu X, Lv X, Zhang L, Li SS, Jin X. Noncoding RNA Lipotherapeutics: A Promising Breakthrough in Pulmonary Hypertension Treatment. Curr Pharm Biotechnol 2025; 26:9-16. [PMID: 38561610 DOI: 10.2174/0113892010302590240321073509] [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: 01/06/2024] [Revised: 03/03/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Pulmonary Hypertension (PH) is a complex cardiovascular disorder characterized by elevated blood pressure in the pulmonary arteries. Current therapeutic approaches for PH have limitations in addressing the underlying molecular mechanisms. This article explores the potential of noncoding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), delivered through Lipid-Based Nanoparticles (LNPs) as a novel treatment strategy. These ncRNAs play critical roles in regulating vascular function and are implicated in PH pathogenesis. LNPs provide a promising method for the efficient and targeted delivery of ncRNAs. Advances in LNP technology, including the incorporation of R8 peptide modification, have shown promise in enhancing the delivery and efficacy of ncRNAs in PH models. Challenges such as biocompatibility, toxicity, and precise targeting must be addressed as these therapies move toward clinical application. The potential of personalized medicine and the integration of artificial intelligence in LNP design are discussed as prospects. In conclusion, ncRNA lipotherapeutics delivered via LNPs offer a transformative approach to treating PH, potentially leading to more effective management and improved patient outcomes in the future. However, continued research and clinical trials are necessary to fully realize their therapeutic potential in the field of PH treatment.
Collapse
Affiliation(s)
- Xuanyi Hu
- School of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Xinxin Lv
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lingzhu Zhang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Shan-Shan Li
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
28
|
Hei B, Zhang A, Yang M, Jiang Y, Guan Z. Liver Kinase B1 Protects Against Hypoxia-Induced Pulmonary Arterial Endothelial Cell Dysfunction via the AMP-Activated Protein Kinase Pathway. Biochem Genet 2024:10.1007/s10528-024-11007-w. [PMID: 39720983 DOI: 10.1007/s10528-024-11007-w] [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: 10/16/2024] [Accepted: 12/15/2024] [Indexed: 12/26/2024]
Abstract
Pulmonary hypertension (PH) is a progressive disease characterized by vascular reHypoxiaing, endothelial cell dysfunction, and inflammation. Liver Kinase B1 (LKB1, also known as STK11) is a central regulator of cell polarity and energy homeostasis. However, its specific role and mechanism of action in PH remain unclear. Human pulmonary arterial endothelial cells (hPAECs) were cultured in vitro to establish PH cell Hypoxias under normoxic and hypoxic conditions. The expression of LKB1 was detected by reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and western blotting, and its effect on hPAECs function was investigated by overexpression and inhibition of LKB1. Furthermore, cell proliferation was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, apoptosis was measured by flow cytometry, inflammatory cytokine secretion was evaluated using enzyme-linked immunosorbent assay (ELISA), and the expression of AMP-activated protein kinase (AMPK) signaling pathway-related proteins was analyzed by western blotting. LKB1 expression was significantly reduced in hypoxia-treated hPAECs compared with that in normoxic controls, and LKB1 overexpression significantly ameliorated the hypoxia-induced decrease in cell proliferation and increase in apoptosis as well as inflammatory factor secretion. The AMPK agonist (GSK621) reversed the dysfunction caused by LKB1 inhibition, indicating that LKB1 regulates hPAECs function through the AMPK signaling pathway. LKB1 plays a protective role in PH by inhibiting hPAECs dysfunction via activation of the AMPK pathway.
Collapse
Affiliation(s)
- Bingchang Hei
- Intensive Care Unit, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161099, China
| | - Anzhe Zhang
- Intensive Care Unit, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161099, China
| | - Meiming Yang
- Respiratory Medicine, Tailai County People's Hospital, Qiqihar, 162400, China
| | - Yunfei Jiang
- Respiratory Medicine Ward Three, The Third Affiliated Hospital of Qiqihar Medical University, No. 3 Taishun Road, Qiqihar, 161099, China
| | - Zhanjiang Guan
- Intensive Care Unit, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, 161099, China.
| |
Collapse
|
29
|
Tan JS, Wei Y, Chong L, Yang Y, Hu S, Wang Y. SGLT2 inhibitors as a potential therapeutic option for pulmonary hypertension: mechanisms and clinical perspectives. Crit Rev Clin Lab Sci 2024; 61:709-725. [PMID: 38847284 DOI: 10.1080/10408363.2024.2361012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/29/2024] [Accepted: 05/24/2024] [Indexed: 11/27/2024]
Abstract
Pulmonary arterial hypertension (PAH), one subtype of pulmonary hypertension (PH), is a life-threatening condition characterized by pulmonary arterial remodeling, elevated pulmonary vascular resistance, and blood pressure in the pulmonary arteries, leading to right heart failure and increased mortality. The disease is marked by endothelial dysfunction, vasoconstriction, and vascular remodeling. The role of Sodium-Glucose Co-Transporter-2 (SGLT2) inhibitors, a class of medications originally developed for diabetes management, is increasingly being explored in the context of cardiovascular diseases, including PAH, due to their potential to modulate these pathophysiological processes. In this review, we systematically examine the burgeoning evidence from both basic and clinical studies that describe the effects of SGLT2 inhibitors on cardiovascular health, with a special emphasis on PAH. By delving into the complex interactions between these drugs and the potential pathobiology that underpins PH, this study seeks to uncover the mechanistic underpinnings that could justify the use of SGLT2 inhibitors as a novel therapeutic approach for PAH. We collate findings that illustrate how SGLT2 inhibitors may influence the normal function of pulmonary arteries, possibly alleviating the pathological hallmarks of PAH such as inflammation, oxidative stress, aberrant cellular proliferation, and so on. Our review thereby outlines a potential paradigm shift in PAH management, suggesting that these inhibitors could play a crucial role in modulating the disease's progression by targeting the potential dysfunctions that drive it. This comprehensive synthesis of existing research underscores the imperative need for further clinical trials to validate the efficacy of SGLT2 inhibitors in PAH and to integrate them into the therapeutic agents used against this challenging disease.
Collapse
Affiliation(s)
- Jiang-Shan Tan
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yixiao Wei
- Peking University Health Science Center, Beijing, China
| | - Lingtao Chong
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanmin Yang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Song Hu
- Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yimeng Wang
- Emergency Center, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease of China, National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Ge Q, Zhang T, Yu J, Lu X, Xiao S, Zhang T, Qing T, Xiao Z, Zeng L, Luo L. A new perspective on targeting pulmonary arterial hypertension: Programmed cell death pathways (Autophagy, Pyroptosis, Ferroptosis). Biomed Pharmacother 2024; 181:117706. [PMID: 39581144 DOI: 10.1016/j.biopha.2024.117706] [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: 05/05/2024] [Revised: 11/10/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe cardiovascular disease characterized by elevated pulmonary vascular resistance, progressive increases in pulmonary artery pressures, ultimately leading to right-sided heart failure, and potentially mortality. Pulmonary vascular remodeling is pivotal in PAH onset and progression. While targeted drug therapies have notably ameliorated PAH prognosis, current medications primarily focus on vascular vasodilation, with limited ability to reverse pulmonary vascular remodeling fundamentally, resulting in suboptimal patient prognoses. Cellular death in pulmonary vasculature, once thought to be confined to apoptosis and necrosis, has evolved with the identification of pyroptosis, autophagy, and ferroptosis, revealing their association with vascular injury in PAH. These novel forms of regulated cellular death impact reactive oxygen species (ROS) generation, calcium stress, and inflammatory cascades, leading to pulmonary vascular cell loss, exacerbating vascular injury, and mediating adverse remodeling, inflammation, immune anomalies, and current emerging mechanisms (such as endothelial-mesenchymal transition, abnormal energy metabolism, and epigenetic regulation) in the pathogenesis of PAH. This review comprehensively delineates the roles of autophagy, pyroptosis, and ferroptosis in PAH, elucidating recent advances in their involvement and regulation of vascular injury. It juxtaposes their distinct functions in PAH and discusses the interplay of these programmed cell deaths in pulmonary vascular injury, highlighting the benefits of combined targeted therapies in mitigating pulmonary arterial hypertension-induced vascular injury, providing novel insights into targeted treatments for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Qingliang Ge
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tianqing Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Jiangbiao Yu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Xuelin Lu
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Sijie Xiao
- Department of Ultrasound, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Ting Zhang
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Tao Qing
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Zhenni Xiao
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China
| | - Liuting Zeng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Li Luo
- Department of Cardiology, Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde City, China.
| |
Collapse
|
31
|
Lyu TT, Wang JY, Tan JS, Yang YM, Wang YM, Zhao J, Qing P, Wu LM, Wang XJ. Causal associations between telomere length and pulmonary arterial hypertension: A two-sample Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40407. [PMID: 39809206 PMCID: PMC11596709 DOI: 10.1097/md.0000000000040407] [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: 08/19/2024] [Accepted: 10/17/2024] [Indexed: 01/16/2025] Open
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening condition characterized by elevated pulmonary artery pressure, leading to right heart failure, and mortality. The role of telomere length, a marker of biological aging, in PAH remains unclear. We utilized summary-level data from genome-wide association studies for various measures of telomere length and PAH. Single nucleotide polymorphisms associated with telomere length at a genome-wide significance level were used as instrumental variables. The inverse variance weighted method was the primary analysis, with sensitivity analyses including the weighted median and Mendelian randomization-Egger regression. The odds ratios and 95% confidence intervals (CI) were calculated to estimate the causal effect of telomere length on PAH risk. The Mendelian randomization analyses revealed no significant causal association between overall telomere length and PAH (odds ratios per standard deviation increase = 1.229, 95% CI: 0.469-3.222, P = .676). Similar null findings were observed for granulocyte, lymphocyte, naive T-cell, memory T-cell, B-cell, and natural killer-cell telomere lengths. Sensitivity analyses confirmed the robustness of the results, with no evidence of horizontal pleiotropy or significant influence of individual single nucleotide polymorphisms on the overall estimates. This Mendelian randomization study didn't support a causal association between telomere length and PAH.
Collapse
Affiliation(s)
- Ting-Ting Lyu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jing-Yang Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Emergency and Critical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiang-Shan Tan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Emergency and Critical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan-Min Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Emergency and Critical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi-Meng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Emergency and Critical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ping Qing
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Emergency and Critical Care Center, Fuwai Hospital, National Center for Cardiovascular Diseases of China, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ling-Min Wu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiao-Jian Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, National Clinical Research Center of Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Fuwai Central-China Hospital, Central-China Branch of National Center for Cardiovascular Diseases, Zhengzhou, China
- Key Laboratory of Pulmonary Vascular Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Fuwai Yunnan Cardiovascular Hospital, Kunming, China
| |
Collapse
|
32
|
Zeder K. Being on Time in Pulmonary Arterial Hypertension: Early Diagnosis in High-Risk Populations. Chest 2024; 166:916-918. [PMID: 39521539 DOI: 10.1016/j.chest.2024.07.139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 11/16/2024] Open
Affiliation(s)
- Katarina Zeder
- Division of Cardiovascular Medicine, School of Medicine, Baltimore, MD; University of Maryland, Institute for Health Computing, Bethesda, MD; Division of Pulmonology, Medical University of Graz, and Ludwig Boltzmann Institute for Lung Vascular Research Graz, Austria.
| |
Collapse
|
33
|
Aun JA, Kwong R, Weber B. Cardiac MRI in Rheumatic Disease. Rheum Dis Clin North Am 2024; 50:735-756. [PMID: 39415377 PMCID: PMC11487115 DOI: 10.1016/j.rdc.2024.07.010] [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] [Indexed: 10/18/2024]
Abstract
Immune-mediated systemic inflammatory disorders present a latent threat for cardiovascular disease. Early involvement may be associated with constitutional symptoms, while clinical evidence of disease may manifest later in an insidious manner. Multimodality imaging is crucial to detect myocardial involvement, with transthoracic echocardiogram as a first-line imaging modality; however, cardiac MRI (CMRI) has the potential to significantly impact our diagnostic and therapeutic approaches through high-fidelity chamber quantification and parametric mapping techniques. Novel imaging techniques are currently under investigation, including stress CMRI, feature tracking CMR, late gadolinium enhancement (LGE) entropy, and 4 dimensional flow CMRI.
Collapse
Affiliation(s)
- Jonathan A Aun
- Heart and Vascular Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Medicine, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD, USA. https://twitter.com/jonathan_aun
| | - Raymond Kwong
- Heart and Vascular Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brittany Weber
- Heart and Vascular Center, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Yan Q, Li P, Liu S, Sun Y, Chen C, Long J, Lin Y, Liang J, Wang H, Zhang L, Wang H, Wang H, Yang S, Lin M, Liu X, Yao J, Tian Z, Chen N, Yang Y, Ai Q. Dihydromyricetin treats pulmonary hypertension by modulating CKLF1/CCR5 axis-induced pulmonary vascular cell pyroptosis. Biomed Pharmacother 2024; 180:117614. [PMID: 39461017 DOI: 10.1016/j.biopha.2024.117614] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024] Open
Abstract
Pulmonary hypertension (PH) is a progressive cardiopulmonary disease characterized by elevated pulmonary artery pressure and vascular remodeling, resulting in poor prognosis and increased mortality rates. Chemokine-like factor 1 (CKLF1) plays a significant role in inducing inflammation and cell proliferation, both of which are critical processes in the pathogenesis of various diseases. Dihydromyricetin (DMY) has garnered attention for its potent anti-inflammatory properties. This study evaluated the protective effects of DMY against PH, demonstrating that DMY treatment can mitigate pyroptosis in pulmonary artery endothelial cells (PAECs) and pulmonary artery smooth muscle cells (PASMCs) in vivo via the CKLF1/CCR5 axis. Results indicated significant improvements in hemodynamics, inflammatory responses, fibrosis, vascular remodeling, and right ventricular hypertrophy in PH rats following DMY treatment. Furthermore, the interaction between CKLF1 and CCR5 was investigated in CKLF1-/- rats after PH induction. DMY was found to downregulate CKLF1 expression and the inflammatory response in the lungs, with its therapeutic efficacy diminished following CKLF1 knockdown. This study underscores the therapeutic potential of DMY in the management of PH and lays a foundation for future research and clinical applications.
Collapse
MESH Headings
- Animals
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Receptors, CCR5/metabolism
- Flavonols/pharmacology
- Flavonols/therapeutic use
- Male
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pyroptosis/drug effects
- Rats, Sprague-Dawley
- Rats
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- MARVEL Domain-Containing Proteins/metabolism
- Vascular Remodeling/drug effects
- Signal Transduction/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Disease Models, Animal
- Cells, Cultured
Collapse
Affiliation(s)
- Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ping Li
- Technology Innovation Center, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal&Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinping Liang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hanlong Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ling Zhang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Hongbin Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huiqin Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuan Liu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jiao Yao
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhifeng Tian
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
35
|
Zeder K. Beyond the Lung: Viewing Treatment Response through the Liver in Pulmonary Arterial Hypertension. Am J Respir Crit Care Med 2024; 210:976-978. [PMID: 38913580 PMCID: PMC11531097 DOI: 10.1164/rccm.202405-1017ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024] Open
Affiliation(s)
- Katarina Zeder
- Division of Cardiovascular Medicine University of Maryland School of Medicine Baltimore, Maryland
- University of Maryland Institute for Health Computing Bethesda, Maryland
- Division of Pulmonology Medical University of Graz Graz, Austria
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Austria
| |
Collapse
|
36
|
Zhang B, Guan Y, Zeng D, Wang R. Arginine methylation and respiratory disease. Transl Res 2024; 272:140-150. [PMID: 38453053 DOI: 10.1016/j.trsl.2024.03.002] [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: 01/04/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Arginine methylation, a vital post-translational modification, plays a pivotal role in numerous cellular functions such as signal transduction, DNA damage response and repair, regulation of gene transcription, mRNA splicing, and protein interactions. Central to this modification is the role of protein arginine methyltransferases (PRMTs), which have been increasingly recognized for their involvement in the pathogenesis of various respiratory diseases. This review begins with an exploration of the biochemical underpinnings of arginine methylation, shedding light on the intricate molecular regulatory mechanisms governed by PRMTs. It then delves into the impact of arginine methylation and the dysregulation of arginine methyltransferases in diverse pulmonary disorders. Concluding with a focus on the therapeutic potential and recent advancements in PRMT inhibitors, this article aims to offer novel perspectives and therapeutic avenues for the management and treatment of respiratory diseases.
Collapse
Affiliation(s)
- Binbin Zhang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, PR China
| | - Youhong Guan
- Department of Infectious Diseases, Hefei Second People's Hospital, Hefei 230001, Anhui Province, PR China
| | - Daxiong Zeng
- Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou 215006, Jiangsu Province, PR China.
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, PR China.
| |
Collapse
|
37
|
Shao Y, Xu J, Chen W, Hao M, Liu X, Zhang R, Wang Y, Dong Y. miR-135b: An emerging player in cardio-cerebrovascular diseases. J Pharm Anal 2024; 14:100997. [PMID: 39211791 PMCID: PMC11350494 DOI: 10.1016/j.jpha.2024.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 09/04/2024] Open
Abstract
miR-135 is a highly conserved miRNA in mammals and includes miR-135a and miR-135b. Recent studies have shown that miR-135b is a key regulatory factor in cardio-cerebrovascular diseases. It is involved in regulating the pathological process of myocardial infarction, myocardial ischemia/reperfusion injury, cardiac hypertrophy, atrial fibrillation, diabetic cardiomyopathy, atherosclerosis, pulmonary hypertension, cerebral ischemia/reperfusion injury, Parkinson's disease, and Alzheimer's disease. Obviously, miR-135b is an emerging player in cardio-cerebrovascular diseases and is expected to be an important target for the treatment of cardio-cerebrovascular diseases. However, the crucial role of miR-135b in cardio-cerebrovascular diseases and its underlying mechanism of action has not been reviewed. Therefore, in this review, we aimed to comprehensively summarize the role of miR-135b and the signaling pathway mediated by miR-135b in cardio-cerebrovascular diseases. Drugs targeting miR-135b for the treatment of diseases and related patents, highlighting the importance of this target and its utility as a therapeutic target for cardio-cerebrovascular diseases, have been discussed.
Collapse
Affiliation(s)
- Yingchun Shao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Wujun Chen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Yanhong Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Yinying Dong
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| |
Collapse
|
38
|
Lu M, Baima YJ, Ni Z, Yang L, Zhang SS, Zhang YT. Advances in the potential of nebulized inhalation for the treatment of pulmonary arterial hypertension. Curr Probl Cardiol 2024; 49:102752. [PMID: 39059783 DOI: 10.1016/j.cpcardiol.2024.102752] [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: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Pulmonary hypertension is a pathophysiologic manifestation of a heterogeneous group of diseases, with the main pathophysiologic mechanisms being persistent pulmonary vasoconstriction and irreversible vascular remodeling. The impact significantly affects the prognosis of patients with pulmonary hypertension. If it is not treated and intervened in time, it may lead to right ventricular failure and further endanger the patient's life. Within the past decade or so, nebulized inhalation therapy is considered to have advantages in the treatment of pulmonary hypertension as a safe, limited, and rapid therapy, for example, inhaled vasodilators (prostate analogs, nitroglycerin, carbon monoxide analogs sildenafil, and nitroprusside), inhaled anti-inflammatory and antiproliferative agents (simvastatin, and selatinib), and inhaled peroxides (levocetirizine) have been recognized as emerging therapeutic approaches in the treatment of pulmonary hypertension as emerging therapeutic approaches. Therefore, this article provides a brief review of recent advances in the potential of nebulized inhaled vasodilators, anti-inflammatory and antiproliferative agents, and anti-peroxides for the treatment of pulmonary hypertension, with the aim of providing different therapeutic options for the treatment of pulmonary hypertension, enhancing the quality of survival, alleviating symptoms, and improving the prognosis of patients with this condition.
Collapse
Affiliation(s)
- Miao Lu
- Tibet University Medical College, Lhasa, Tibet Autonomous Region 850000, China; Department of Pulmonary Medicine, Lhasa People's Hospital, Lhasa, Tibet Autonomous Region 850013, China
| | - Yang-Jin Baima
- Department of Pulmonary Medicine, Lhasa People's Hospital, Lhasa, Tibet Autonomous Region 850013, China
| | - Zhu Ni
- Department of Pulmonary Medicine, Lhasa People's Hospital, Lhasa, Tibet Autonomous Region 850013, China
| | - Li Yang
- Department of Pulmonary Medicine, Lhasa People's Hospital, Lhasa, Tibet Autonomous Region 850013, China
| | - Song-Shan Zhang
- Tibet University Medical College, Lhasa, Tibet Autonomous Region 850000, China; Department of External Medicine, Tibet Autonomous Region People's Hospital, Lhasa, Tibet Autonomous Region 850000, China
| | - Yun-Tao Zhang
- Department of Pulmonary Medicine, Lhasa People's Hospital, Lhasa, Tibet Autonomous Region 850013, China.
| |
Collapse
|
39
|
Chen Z, Song L, Zhong M, Pang L, Sun J, Xian Q, Huang T, Xie F, Cheng J, Fu K, Huang Z, Guo D, Chen R, Sun X, Huang C. A comprehensive analysis of genes associated with hypoxia and cuproptosis in pulmonary arterial hypertension using machine learning methods and immune infiltration analysis: AHR is a key gene in the cuproptosis process. Front Med (Lausanne) 2024; 11:1435068. [PMID: 39391037 PMCID: PMC11464361 DOI: 10.3389/fmed.2024.1435068] [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: 05/19/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Background Pulmonary arterial hypertension (PAH) is a serious condition characterized by elevated pulmonary artery pressure, leading to right heart failure and increased mortality. This study investigates the link between PAH and genes associated with hypoxia and cuproptosis. Methods We utilized expression profiles and single-cell RNA-seq data of PAH from the GEO database and genecad. Genes related to cuproptosis and hypoxia were identified. After normalizing the data, differential gene expression was analyzed between PAH and control groups. We performed clustering analyses on cuproptosis-related genes and constructed a weighted gene co-expression network (WGCNA) to identify key genes linked to cuproptosis subtype scores. KEGG, GO, and DO enrichment analyses were conducted for hypoxia-related genes, and a protein-protein interaction (PPI) network was created using STRING. Immune cell composition differences were examined between groups. SingleR and Seurat were used for scRNA-seq data analysis, with PCA and t-SNE for dimensionality reduction. We analyzed hub gene expression across single-cell clusters and built a diagnostic model using LASSO and random forest, optimizing parameters with 10-fold cross-validation. A total of 113 combinations of 12 machine learning algorithms were employed to evaluate model accuracy. GSEA was utilized for pathway enrichment analysis of AHR and FAS, and a Nomogram was created to assess risk impact. We also analyzed the correlation between key genes and immune cell types using Spearman correlation. Results We identified several diagnostic genes for PAH linked to hypoxia and cuproptosis. PPI networks illustrated relationships among these hub genes, with immune infiltration analysis highlighting associations with monocytes, macrophages, and CD8 T cells. The genes AHR, FAS, and FGF2 emerged as key markers, forming a robust diagnostic model (NaiveBayes) with an AUC of 0.9. Conclusion AHR, FAS, and FGF2 were identified as potential biomarkers for PAH, influencing cell proliferation and inflammatory responses, thereby offering new insights for PAH prevention and treatment.
Collapse
Affiliation(s)
- Zuguang Chen
- Central People’s Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| | - Lingyue Song
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ming Zhong
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lingpin Pang
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jie Sun
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qian Xian
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Tao Huang
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Fengwei Xie
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junfen Cheng
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Kaili Fu
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zhihai Huang
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Dingyu Guo
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Riken Chen
- Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Xishi Sun
- Emergency Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Chunyi Huang
- Central People’s Hospital of Zhanjiang, Zhanjiang, Guangdong, China
| |
Collapse
|
40
|
Kudryashova TV, Zaitsev SV, Jiang L, Buckley BJ, McGuckin JP, Goncharov D, Zhyvylo I, Lin D, Newcomb G, Piper B, Bogamuwa S, Saiyed A, Teos L, Pena A, Ranson M, Greenland JR, Wolters PJ, Kelso MJ, Poncz M, DeLisser HM, Cines DB, Goncharova EA, Farkas L, Stepanova V. PAI-1 deficiency drives pulmonary vascular smooth muscle remodeling and pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2024; 327:L319-L326. [PMID: 38860847 PMCID: PMC11444499 DOI: 10.1152/ajplung.00110.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/30/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease characterized by vasoconstriction and remodeling of small pulmonary arteries (PAs). Central to the remodeling process is a switch of pulmonary vascular cells to a proliferative, apoptosis-resistant phenotype. Plasminogen activator inhibitors-1 and -2 (PAI-1 and PAI-2) are the primary physiological inhibitors of urokinase-type and tissue-type plasminogen activators (uPA and tPA), but their roles in PAH are unsettled. Here, we report that: 1) PAI-1, but not PAI-2, is deficient in remodeled small PAs and in early-passage PA smooth muscle and endothelial cells (PASMCs and PAECs) from subjects with PAH compared with controls; 2) PAI-1-/- mice spontaneously develop pulmonary vascular remodeling associated with upregulation of mTORC1 signaling, pulmonary hypertension (PH), and right ventricle (RV) hypertrophy; and 3) pharmacological inhibition of uPA in human PAH PASMCs suppresses proproliferative mTORC1 and SMAD3 signaling, restores PAI-1 levels, reduces proliferation, and induces apoptosis in vitro, and prevents the development of SU5416/hypoxia-induced PH and RV hypertrophy in vivo in mice. These data strongly suggest that downregulation of PAI-1 in small PAs promotes vascular remodeling and PH due to unopposed activation of uPA and consequent upregulation of mTOR and transforming growth factor-β (TGF-β) signaling in PASMCs, and call for further studies to determine the potential benefits of targeting the PAI-1/uPA imbalance to attenuate and/or reverse pulmonary vascular remodeling and PH.NEW & NOTEWORTHY This study identifies a novel role for the deficiency of plasminogen activator inhibitor (PAI)-1 and resultant unrestricted uPA activity in PASMC remodeling and PH in vitro and in vivo, provides novel mechanistic link from PAI-1 loss through uPA-induced Akt/mTOR and TGFβ-Smad3 upregulation to pulmonary vascular remodeling in PH, and suggests that inhibition of uPA to rebalance the uPA-PAI-1 tandem might provide a novel approach to complement current therapies used to mitigate this pulmonary vascular disease.
Collapse
MESH Headings
- Animals
- Plasminogen Activator Inhibitor 1/metabolism
- Plasminogen Activator Inhibitor 1/genetics
- Vascular Remodeling
- Humans
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Signal Transduction
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cell Proliferation
- Mice, Knockout
- Mechanistic Target of Rapamycin Complex 1/metabolism
- Mice, Inbred C57BL
- Apoptosis
- Urokinase-Type Plasminogen Activator/metabolism
- Urokinase-Type Plasminogen Activator/genetics
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Plasminogen Activator Inhibitor 2/metabolism
- Plasminogen Activator Inhibitor 2/genetics
Collapse
Grants
- R01 HL159256 NHLBI NIH HHS
- R01HL150638 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL139881 NHLBI NIH HHS
- Nina Ireland Program for Lung Health UCSF | Department of Medicine, University of California, San Francisco (UCSF Department of Medicine)
- R01 HL141462 NHLBI NIH HHS
- R01 HL166932 NHLBI NIH HHS
- R35HL150698 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R24 HL123767 NHLBI NIH HHS
- I01 CX002011 CSRD VA
- R01 HL130261 NHLBI NIH HHS
- R01HL130261 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL166932 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- LAM0139P07-19 LAM Foundation (TheLAMFoundation)
- R01HL139881 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- APP1181179 DHAC | National Health and Medical Research Council (NHMRC)
- TS150032 DOD | USA | MEDCOM | MRDC | U.S. Army Medical Research Acquisition Activity (USAMRAA)
- CX002011 ORD | Clinical Science Research and Development (CSRD)
- Cardiovascular Medical Research and Education Fund (CMREF)
- RO1HL159256 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL172488 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL150638 NHLBI NIH HHS
- R01 HL172488 NHLBI NIH HHS
- RO1HL141462 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R35 HL150698 NHLBI NIH HHS
Collapse
Affiliation(s)
- Tatiana V Kudryashova
- University of Pittsburgh Heart, Blood, and Vascular Medicine Institute, Pittsburgh, Pennsylvania, United States
| | - Sergei V Zaitsev
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Lifeng Jiang
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Benjamin J Buckley
- School of Chemistry and Molecular Bioscience and Molecular Horizons Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Joshua P McGuckin
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, United States
| | - Dmitry Goncharov
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Iryna Zhyvylo
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Derek Lin
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Geoffrey Newcomb
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Bryce Piper
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Srimathi Bogamuwa
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Aisha Saiyed
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Leyla Teos
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Andressa Pena
- University of Pittsburgh Heart, Blood, and Vascular Medicine Institute, Pittsburgh, Pennsylvania, United States
| | - Marie Ranson
- School of Chemistry and Molecular Bioscience and Molecular Horizons Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - John R Greenland
- School of Medicine, University of California, San Francisco, California, United States
- San Francisco Veterans Affairs Health Care System, San Francisco, California, United States
| | - Paul J Wolters
- School of Medicine, University of California, San Francisco, California, United States
| | - Michael J Kelso
- School of Chemistry and Molecular Bioscience and Molecular Horizons Institute, University of Wollongong, Wollongong, New South Wales, Australia
| | - Mortimer Poncz
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Horace M DeLisser
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Douglas B Cines
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Elena A Goncharova
- Department of Internal Medicine, University of California, Davis, California, United States
| | - Laszlo Farkas
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Victoria Stepanova
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
41
|
Zhang S, Li S, Gao L, Zhao Q, Yang T, Zeng Q, Huang Z, Li X, Duan A, Wang Y, Zhao Z, Luo Q, Liu Z. Effects of malnutrition on disease severity and adverse outcomes in idiopathic pulmonary arterial hypertension: a retrospective cohort study. Respir Res 2024; 25:292. [PMID: 39080722 PMCID: PMC11290113 DOI: 10.1186/s12931-024-02925-9] [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: 05/22/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Malnutrition is common in patients with chronic cardiovascular disease and is associated with significantly higher all-cause mortality. Approximately one-third of patients with heart failure are malnourished. However, the relationship between malnutrition and idiopathic pulmonary arterial hypertension (IPAH) remains unclear. This study aimed to clarify the prognostic value of malnutrition in patients with IPAH. METHODS A total of 432 consecutive participants with IPAH were included in this study between March 2013 and August 2021. Three common malnutrition assessment tools, including the geriatric nutritional risk index (GNRI), prognostic nutritional index (PNI), and controlling nutritional status (CONUT) score, were used to evaluate the nutritional status of patients with IPAH. The relationships between the malnutrition tools and long-term adverse outcomes were determined using restricted cubic splines and multivariate Cox regression models. RESULTS During a mean follow-up of 3.1 years, 158 participants experienced clinical worsening or all-cause death. Patients were stratified into the low-, intermediate- and high-risk groups based on the European Society of Cardiology (ESC) risk stratification, and the PNI (55.9 ± 5.7 vs. 54.4 ± 7.2 vs. 51.1 ± 7.1, P = 0.005) and CONUT score (2.1 ± 0.9 vs. 2.5 ± 1.2 vs. 3.3 ± 1.1, P < 0.001) identified these patient groups better than the GNRI. All three malnutrition tools were associated with well-validated variables that reflected IPAH severity, such as the World Health Organization functional class, 6-min walk distance, and N-terminal pro-brain natriuretic peptide level. The CONUT score exhibited better predictive ability than both the GNRI (ΔAUC = 0.059, P < 0.001) and PNI (ΔAUC = 0.095, P < 0.001) for adverse outcomes and significantly improved reclassification and discrimination beyond the ESC risk score. Multivariable Cox regression analysis indicated that only the CONUT score (hazard ratio = 1.363, 95% confidence interval 1.147, 1.619 per 1.0-standard deviation increment, P < 0.001) independently predicted adverse outcomes. CONCLUSIONS The malnutrition status was associated with disease severity in patients with IPAH. The CONUT score provided additional information regarding the risk of clinically worsening events, making it a meaningful risk stratification tool for these patients.
Collapse
Affiliation(s)
- Sicheng Zhang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Sicong Li
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Luyang Gao
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Qing Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Tao Yang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Qixian Zeng
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zhihua Huang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Xin Li
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Anqi Duan
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yijia Wang
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Zhihui Zhao
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Qin Luo
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| | - Zhihong Liu
- Center for Respiratory and Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 167, Beilishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
42
|
Zhang Z, Yang Z, Wang S, Wang X, Mao J. Decoding ferroptosis: Revealing the hidden assassin behind cardiovascular diseases. Biomed Pharmacother 2024; 176:116761. [PMID: 38788596 DOI: 10.1016/j.biopha.2024.116761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/09/2024] [Accepted: 05/17/2024] [Indexed: 05/26/2024] Open
Abstract
The discovery of regulatory cell death processes has driven innovation in cardiovascular disease (CVD) therapeutic strategies. Over the past decade, ferroptosis, an iron-dependent form of regulated cell death driven by excessive lipid peroxidation, has been shown to drive the development of multiple CVDs. This review provides insights into the evolution of the concept of ferroptosis, the similarities and differences with traditional modes of programmed cell death (e.g., apoptosis, autophagy, and necrosis), as well as the core regulatory mechanisms of ferroptosis (including cystine/glutamate transporter blockade, imbalance of iron metabolism, and lipid peroxidation). In addition, it provides not only a detailed review of the role of ferroptosis and its therapeutic potential in widely studied CVDs such as coronary atherosclerotic heart disease, myocardial infarction, myocardial ischemia/reperfusion injury, heart failure, cardiomyopathy, and aortic aneurysm but also an overview of the phenomenon and therapeutic perspectives of ferroptosis in lesser-addressed CVDs such as cardiac valvulopathy, pulmonary hypertension, and sickle cell disease. This article aims to integrate this knowledge to provide a comprehensive view of ferroptosis in a wide range of CVDs and to drive innovation and progress in therapeutic strategies in this field.
Collapse
Affiliation(s)
- Zeyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhihua Yang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China; Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Shuai Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China
| | - Xianliang Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| | - Jingyuan Mao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin 300381, China.
| |
Collapse
|
43
|
Zhang JJ, Mao-Mao, Shao MM, Wang MC. Therapeutic potential of natural flavonoids in pulmonary arterial hypertension: A review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155535. [PMID: 38537442 DOI: 10.1016/j.phymed.2024.155535] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/06/2024] [Accepted: 03/12/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a fatal disease caused by pulmonary vascular remodeling, with a high incidence and mortality. At present, many clinical drugs for treating PAH mainly exert effects by relaxing the pulmonary artery, with limited therapeutic effects, so the search for viable therapeutic agents continues uninterrupted. In recent years, natural flavonoids have shown promising potential in the treatment of cardiovascular diseases. It is necessary to comprehensively elucidate the potential of natural flavonoids to combat PAH. PURPOSE To evaluate the potential of natural flavonoids to hinder or slow down the occurrence and development of PAH, and to identify promising drug discovery candidates. METHODS Literature was collected from PubMed, Science Direct, Web of science, CNKI databases and Google scholar. The search terms used included "pulmonary arterial hypertension", "pulmonary hypertension", "natural products", "natural flavonoids", "traditional chinese medicine", etc., and several combinations of these keywords. RESULTS The resources, structural characteristics, mechanisms, potential and prospect strategies of natural flavonoids for treating PAH were summarized. Natural flavonoids offer different solutions as possible treatments for PAH. These mechanisms may involve various pathways and molecular targets related to the pathogenesis of PAH, such as inflammation, oxidative stress, vascular remodeling, genetic, ion channels, cell proliferation and autophagy. In addition, prospect strategies of natural flavonoids for anti-PAH including structural modification and nanomaterial delivery systems have been explored. This review suggests that the potential of natural flavonoids as alternative therapeutic agents in the prevention and treatment of PAH holds promise for future research and clinical applications. CONCLUSION Despite displaying the enormous potential of flavonoids in PAH, some limitations need to be further explored. Firstly, using advanced drug discovery tools, including computer-aided design and high-throughput screening, to further investigate the safety, biological activity, and precise mechanism of action of flavonoids. Secondly, exploring the structural modifications of these compounds is expected to optimize their efficacy. Lastly, it is necessary to conduct well controlled clinical trials and a comprehensive evaluation of potential side effects to determine their effectiveness and safety.
Collapse
Affiliation(s)
- Jin-Jing Zhang
- Department of pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, China
| | - Mao-Mao
- Department of pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, China
| | - Min-Min Shao
- Department of pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, China
| | - Meng-Chuan Wang
- Department of pharmacy, Affiliated Cixi Hospital, Wenzhou Medical University, China.
| |
Collapse
|
44
|
Yuan Y, Li S, Yan M, Yang Y, Zhong C, Hu Y. Genetically determined gut microbiota associates with pulmonary arterial hypertension: a Mendelian randomization study. BMC Pulm Med 2024; 24:235. [PMID: 38745167 PMCID: PMC11094871 DOI: 10.1186/s12890-024-02877-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/24/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Emerging evidences have demonstrated that gut microbiota composition is associated with pulmonary arterial hypertension (PAH). However, the underlying causality between intestinal dysbiosis and PAH remains unresolved. METHOD An analysis using the two-sample Mendelian randomization (MR) approach was conducted to examine the potential causal relationship between gut microbiota and PAH. To assess exposure data, genetic variants associated with 196 bacterial traits were extracted from the MiBioGen consortium, which included a sample size of 18,340 individuals. As for the outcomes, summary statistics for PAH were obtained from the NHGRI-EBI GWAS Catalog, which conducted a meta-analysis of four independent studies comprising a total of 11,744 samples. Causal effects were estimated employing various methods, including inverse variance weighted (IVW), MR-Egger, weighted median, weight mode and simple mode, with sensitivity analyses also being implemented with Cochran's Q test, MR-Egger intercept test, MR-PRESSO, leave-one-out analysis, and funnel plots. RESULTS Following false discovery rate (FDR) correction, the genetically predicted genus Eubacterium fissicatena group (odds ratio (OR) 1.471, 95% confidence interval (CI) 1.178-1.837, q = 0.076) exhibited a causal association with PAH. In addition, the genus LachnospiraceaeUCG004 (OR 1.511, 95% CI 1.048-2.177) and genus RuminococcaceaeUCG002 (OR 1.407, 95% CI 1.040-1.905) showed a suggestive increased risk of PAH, while genus Eubacterium eligens group (OR 0.563, 95% CI 0.344-0.922), genus Phascolarctobacterium (OR 0.692, 95% CI 0.487-0.982), genus Erysipelatoclostridium (OR 0.757, 95% CI 0.579-0.989) and genus T-yzzerella3 (OR 0.768, 95% CI 0.624-0.945) were found to have nominal protective effect against PAH. CONCLUSION The findings from our MR study have revealed a potential causal relationship between gut microbiota and PAH. Specifically, we have identified four types of gut microbiota that exhibit a protective effect on PAH, as well as three types that have a detrimental impact on PAH, thereby offering valuable insights for future mechanistic and clinical investigations in the field of PAH.
Collapse
Affiliation(s)
- Ye Yuan
- Department of Cardiovascular Surgery, Daping Hospital, Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Shan Li
- Department of Hepatobiliary and Pancreatic Tumor Center, Chongqing University Cancer Hospital, 181, Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Manrong Yan
- Department of Cardiovascular Surgery, Daping Hospital, Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Yan Yang
- Department of Cardiovascular Surgery, Daping Hospital, Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Changming Zhong
- Department of Cardiovascular Surgery, Daping Hospital, Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Yijie Hu
- Department of Cardiovascular Surgery, Daping Hospital, Army Medical University, No.10 Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
45
|
Yang Y, Zhang H, Wang Y, Xu J, Shu S, Wang P, Ding S, Huang Y, Zheng L, Yang Y, Xiong C. Promising dawn in the management of pulmonary hypertension: The mystery veil of gut microbiota. IMETA 2024; 3:e159. [PMID: 38882495 PMCID: PMC11170974 DOI: 10.1002/imt2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 11/25/2023] [Indexed: 06/18/2024]
Abstract
The gut microbiota is a complex community of microorganisms inhabiting the intestinal tract, which plays a vital role in human health. It is intricately involved in the metabolism, and it also affects diverse physiological processes. The gut-lung axis is a bidirectional pathway between the gastrointestinal tract and the lungs. Recent research has shown that the gut microbiome plays a crucial role in immune response regulation in the lungs and the development of lung diseases. In this review, we present the interrelated factors concerning gut microbiota and the associated metabolites in pulmonary hypertension (PH), a lethal disease characterized by elevated pulmonary vascular pressure and resistance. Our research team explored the role of gut-microbiota-derived metabolites in cardiovascular diseases and established the correlation between metabolites such as putrescine, succinate, trimethylamine N-oxide (TMAO), and N, N, N-trimethyl-5-aminovaleric acid with the diseases. Furthermore, we found that specific metabolites, such as TMAO and betaine, have significant clinical value in PH, suggesting their potential as biomarkers in disease management. In detailing the interplay between the gut microbiota, their metabolites, and PH, we underscored the potential therapeutic approaches modulating this microbiota. Ultimately, we endeavor to alleviate the substantial socioeconomic burden associated with this disease. This review presents a unique exploratory analysis of the link between gut microbiota and PH, intending to propel further investigations in the gut-lung axis.
Collapse
Affiliation(s)
- Yicheng Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Hanwen Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Yaoyao Wang
- State Key Laboratory of Cardiovascular Disease, Department of Nephrology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Department of Genetics University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | - Songren Shu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Peizhi Wang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
- Center for Molecular Cardiology University of Zurich Zurich Switzerland
| | - Shusi Ding
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
| | - Yuan Huang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiac Surgery Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Lemin Zheng
- China National Clinical Research Center for Neurological Diseases, Tiantan Hospital, Advanced Innovation Center for Human Brain Protection The Capital Medical University Beijing China
- Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, School of Basic Medical Sciences, Health Science Center The Institute of Cardiovascular Sciences and Institute of Systems Biomedicine, Peking University Beijing China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Changming Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| |
Collapse
|
46
|
Khalil S, Tabowei G, Kaur M, Dadzie SK, Kansakar S, Moqattash M, Komminni PK, Palleti SK. Effect of Pulmonary Hypertension on Survival Outcomes in Patients With Transcatheter Aortic Valve Replacement: A Systematic Review and Meta-Analysis. Cureus 2024; 16:e58540. [PMID: 38957831 PMCID: PMC11218420 DOI: 10.7759/cureus.58540] [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] [Accepted: 04/18/2024] [Indexed: 07/04/2024] Open
Abstract
The aim of this meta-analysis was to determine the effect of pulmonary hypertension (PH) on survival in patients undergoing transcatheter aortic valve replacement (TAVR). The present study was conducted according to the guidelines of Preferred Reporting of Systematic Review and Meta-Analysis (PRISMA). We conducted a comprehensive search of electronic databases including PubMed/MEDLINE, Embase, Cochrane Library, and Web of Science from January 1, 2015, to March 10, 2024. Outcomes assessed in this meta-analysis included early and late all-cause mortality and cardiovascular mortality. Total 15 studies were integrated into the pooled analysis to assess the impact of PH on outcomes among patients undergoing TAVR, comprising a total sample size of 35,732 individuals. The pooled prevalence of PH stood at 52.57% (n=18,767). Predominantly, the studies were conducted in the United States (n=6), followed by Germany (n=3), with one study each from Japan, Italy, Switzerland, Brazil, Poland, and Australia. Pooled analysis showed that risk of short-term mortality was greater in patients with PH compared to patients without PH (risk ratio (RR): 1.46, 95% CI: 1.19 to 1.80). Risk of long-term mortality was greater in patients with PH (RR: 1.42, 95% CI: 1.29 to 1.55). Risk of cardiovascular mortality was also greater in patients with PH compared to patients without PH (RR: 1.66, 95% CI: 1.36 to 2.02). We advocate for further research to address gaps in understanding different types of PH and their impacts on mortality and cardiovascular outcomes.
Collapse
Affiliation(s)
| | - Godfrey Tabowei
- Internal Medicine, Texas Tech University Health Sciences Center, Odessa, USA
| | - Mandeep Kaur
- Internal Medicine, HCA Capital Hospital, Tallahassee, USA
| | - Samuel K Dadzie
- Internal Medicine, Piedmont Athens Regional Medical Center, Athens, USA
| | - Sajog Kansakar
- Internal Medicine, Maimonides Medical Center, Brooklyn, USA
| | | | | | - Sujith K Palleti
- Nephrology, Louisiana State University Health Sciences Center, Shreveport, USA
| |
Collapse
|
47
|
Walker M, Moore H, Ataya A, Pham A, Corris PA, Laubenbacher R, Bryant AJ. A perfectly imperfect engine: Utilizing the digital twin paradigm in pulmonary hypertension. Pulm Circ 2024; 14:e12392. [PMID: 38933181 PMCID: PMC11199193 DOI: 10.1002/pul2.12392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Pulmonary hypertension (PH) is a severe medical condition with a number of treatment options, the majority of which are introduced without consideration of the underlying mechanisms driving it within an individual and thus a lack of tailored approach to treatment. The one exception is a patient presenting with apparent pulmonary arterial hypertension and shown to have vaso-responsive disease, whose clinical course and prognosis is significantly improved by high dose calcium channel blockers. PH is however characterized by a relative abundance of available data from patient cohorts, ranging from molecular data characterizing gene and protein expression in different tissues to physiological data at the organ level and clinical information. Integrating available data with mechanistic information at the different scales into computational models suggests an approach to a more personalized treatment of the disease using model-based optimization of interventions for individual patients. That is, constructing digital twins of the disease, customized to a patient, promises to be a key technology for personalized medicine, with the aim of optimizing use of existing treatments and developing novel interventions, such as new drugs. This article presents a perspective on this approach in the context of a review of existing computational models for different aspects of the disease, and it lays out a roadmap for a path to realizing it.
Collapse
Affiliation(s)
- Melody Walker
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Helen Moore
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Ali Ataya
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Ann Pham
- University of Florida College of MedicineGainesvilleFloridaUSA
| | - Paul A. Corris
- The Faculty of Medical Sciences Newcastle UniversityNewcastle upon TyneUK
| | | | | |
Collapse
|
48
|
Zhao H, Song J, Li X, Xia Z, Wang Q, Fu J, Miao Y, Wang D, Wang X. The role of immune cells and inflammation in pulmonary hypertension: mechanisms and implications. Front Immunol 2024; 15:1374506. [PMID: 38529271 PMCID: PMC10962924 DOI: 10.3389/fimmu.2024.1374506] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/26/2024] [Indexed: 03/27/2024] Open
Abstract
Pulmonary hypertension (PH) is a malignant disease with progressive increase of pulmonary vascular pressure, which eventually leads to right heart failure. More and more evidences show that immune cells and inflammation play an important role in the occurrence and development of PH. In the context of pulmonary vascular diseases, immune cells migrate into the walls of the pulmonary vascular system. This leads to an increase in the levels of cytokines and chemokines in both the bloodstream and the surrounding tissues of the pulmonary vessels. As a result, new approaches such as immunotherapy and anti-inflammatory treatments are being considered as potential strategies to halt or potentially reverse the progression of PH. We reviewed the potential mechanisms of immune cells, cytokines and chemokines in PH development. The potential relationship of vascular cells or bone morphogenetic protein receptor 2 (BMPR2) in immune regulation was also expounded. The clinical application and future prospect of immunotherapy were further discussed.
Collapse
Affiliation(s)
- Hui Zhao
- School of Materials and Chemistry, Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Jialin Song
- Department of Limb Trauma, Wendeng Orthopaedic Hospital of Shandong Province, Weihai, Shandong, China
| | - Xiujun Li
- Department of Medicine, Chifeng University, Chifeng, China
| | - Zhaoyi Xia
- Department of Library, Children's Hospital Affiliated to Shandong University, Jinan, Shandong, China
- Department of Library, Jinan Children's Hospital, Shandong, Jinan, Shandong, China
| | - Qian Wang
- School of Materials and Chemistry, Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Jiaqi Fu
- School of Materials and Chemistry, Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Yuqing Miao
- School of Materials and Chemistry, Institute of Bismuth and Rhenium, University of Shanghai for Science and Technology, Shanghai, China
| | - Dapeng Wang
- Department of Intensive Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu, China
| | - Xuguang Wang
- Department of Limb Trauma, Wendeng Orthopaedic Hospital of Shandong Province, Weihai, Shandong, China
| |
Collapse
|
49
|
Singh N, Al-Naamani N, Brown MB, Long GM, Thenappan T, Umar S, Ventetuolo CE, Lahm T. Extrapulmonary manifestations of pulmonary arterial hypertension. Expert Rev Respir Med 2024; 18:189-205. [PMID: 38801029 PMCID: PMC11713041 DOI: 10.1080/17476348.2024.2361037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
INTRODUCTION Extrapulmonary manifestations of pulmonary arterial hypertension (PAH) may play a critical pathobiological role and a deeper understanding will advance insight into mechanisms and novel therapeutic targets. This manuscript reviews our understanding of extrapulmonary manifestations of PAH. AREAS COVERED A group of experts was assembled and a complimentary PubMed search performed (October 2023 - March 2024). Inflammation is observed throughout the central nervous system and attempts at manipulation are an encouraging step toward novel therapeutics. Retinal vascular imaging holds promise as a noninvasive method of detecting early disease and monitoring treatment responses. PAH patients have gut flora alterations and dysbiosis likely plays a role in systemic inflammation. Despite inconsistent observations, the roles of obesity, insulin resistance and dysregulated metabolism may be illuminated by deep phenotyping of body composition. Skeletal muscle dysfunction is perpetuated by metabolic dysfunction, inflammation, and hypoperfusion, but exercise training shows benefit. Renal, hepatic, and bone marrow abnormalities are observed in PAH and may represent both end-organ damage and disease modifiers. EXPERT OPINION Insights into systemic manifestations of PAH will illuminate disease mechanisms and novel therapeutic targets. Additional study is needed to understand whether extrapulmonary manifestations are a cause or effect of PAH and how manipulation may affect outcomes.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
| | - Nadine Al-Naamani
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Mary Beth Brown
- Department of Rehabilitation Medicine, University of Washington School of Medicine, Seattle, WA
| | - Gary Marshall Long
- Department of Kinesiology, Health and Sport Sciences, University of Indianapolis, Indianapolis, IN
| | - Thenappan Thenappan
- Section of Advanced Heart Failure and Pulmonary Hypertension, Cardiovascular Division, University of Minnesota, Minneapolis, MN
| | - Soban Umar
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Corey E. Ventetuolo
- Department of Medicine, Warren Alpert School of Medicine at Brown University, Providence, RI
- Department of Health Services, Policy and Practice, Brown University, Providence, RI
| | - Tim Lahm
- Department of Medicine, National Jewish Health, Denver, CO
- Department of Medicine, University of Colorado, Aurora, CO
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO
| |
Collapse
|
50
|
Leiva O, Beaty W, Soo S, Agarwal MA, Yang EH. Cancer Therapy-Associated Pulmonary Hypertension and Right Ventricular Dysfunction: Etiologies and Prognostic Implications. Rev Cardiovasc Med 2024; 25:87. [PMID: 39076943 PMCID: PMC11263834 DOI: 10.31083/j.rcm2503087] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 07/31/2024] Open
Abstract
Advances in cancer therapies have improved oncologic outcomes but can potentially expose patients to risk of cardiovascular toxicity. While left ventricular (LV) dysfunction is a well-known cardiotoxicity of cancer therapy. Pulmonary hypertension (PH) and right ventricular (RV) dysfunction are seen with several cancer therapies, including alkylating agents, tyrosine kinase inhibitors (TKIs), and immunotherapy, and are associated with significant morbidity and mortality. Awareness and recognition of cancer therapy-associated PH and RV dysfunction is critical to identify underlying etiologies and institute the appropriate therapy. However, gaps exist in the current literature on the epidemiology of PH and RV dysfunction in cancer, underlying pathophysiology and optimal management strategies.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - William Beaty
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Steven Soo
- Department of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Manyoo A. Agarwal
- Cardio-Oncology Program, Heart, Vascular and Thoracic Institute, Cleveland Clinic Abu Dhabi, 00000 Abu Dhabi, United Arab Emirates
| | - Eric H. Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|