1
|
Gao Z. New insights into Smad3 in cardiac fibrosis. Gene 2025; 952:149418. [PMID: 40089084 DOI: 10.1016/j.gene.2025.149418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Damage to myocardial tissues, leading to myocardial fibrosis, is a significant pathological hallmark across various heart diseases. SMAD3, a central transcriptional regulator within the transforming growth factor-beta (TGF-β) signaling pathway, plays a pivotal role in the pathological progression of myocardial fibrosis and cardiac remodeling. It intricately regulates physiological and pathological processes encompassing cell proliferation, differentiation, tissue repair, and fibrosis. Notably, SMAD3 exerts crucial influences in myocardial fibrosis subsequent to myocardial infarction, pressure overload-induced myocardial fibrosis, diabetic cardiomyopathy (DCM), aging-associated cardiac fibrosis and myocarditis-related myocardial fibrosis. The targeted modulation of genes or the utilization of compounds, including traditional Chinese medicine (paeoniflorin, baicalin, and genistein et al.) and other pharmaceutical agents that modulate SMAD3, may offer avenues for restraining the pathological cascade of myocardial fibrosis. Consequently, targeted regulation of SMAD3 associated with myocardial fibrosis may herald novel therapeutic paradigms for ameliorating myocardial diseases.
Collapse
Affiliation(s)
- Zhen Gao
- Liaocheng Vocational and Technical College, Shandong, China.
| |
Collapse
|
2
|
Shooshtarian AK, O'Gallagher K, Shah AM, Zhang M. SERCA2a dysfunction in the pathophysiology of heart failure with preserved ejection fraction: a direct role is yet to be established. Heart Fail Rev 2025; 30:545-564. [PMID: 39843817 PMCID: PMC11991975 DOI: 10.1007/s10741-025-10487-1] [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] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
With rising incidence, mortality and limited therapeutic options, heart failure with preserved ejection fraction (HFpEF) remains one of the most important topics in cardiovascular medicine today. Characterised by left ventricular diastolic dysfunction partially due to impaired Ca2+ homeostasis, one ion channel in particular, SarcoEndoplasmic Reticulum Ca2+-ATPase (SERCA2a), may play a significant role in its pathophysiology. A better understanding of the complex mechanisms interplaying to contribute to SERCA2a dysfunction will help develop treatments targeting it and thus address the growing clinical challenge HFpEF poses. This review examines the conflicting evidence present for changes in SERCA2a expression and activity in HFpEF, explores potential underlying mechanisms, and finally evaluates the drug and gene therapy trials targeting SERCA2a in heart failure. Recent positive results from trials involving widely used anti-diabetic agents such as sodium-glucose co-transporter protein 2 inhibitors (SGLT2i) and glucagon-like peptide-1 (GLP-1) agonists offer advancement in HFpEF management. The potential interplay between these agents and SERCA2a regulation presents a novel angle that could open new avenues for modulating diastolic function; however, the mechanistic research in this emerging field is limited. Overall, the direct role of SERCA2a dysfunction in HFpEF remains undetermined, highlighting the need for well-designed pre-clinical studies and robust clinical trials.
Collapse
Affiliation(s)
- Adam Kia Shooshtarian
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Kevin O'Gallagher
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK
| | - Min Zhang
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London British Heart Foundation Centre of Research Excellence, London, UK.
| |
Collapse
|
3
|
Bibi A, Madè A, Greco S, Garcia-Manteiga JM, Tascini AS, Tastsoglou S, Zaccagnini G, Leszek P, Gaetano C, Martelli F. Circular PVT1 promotes cardiac fibroblast activation interacting with miR-30a-5p and miR-125b-5p. Cell Death Dis 2025; 16:325. [PMID: 40258819 DOI: 10.1038/s41419-025-07652-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/23/2025]
Abstract
Circular RNAs (circRNAs) are involved in the pathogenesis of several cardiovascular diseases, including heart failure. In this study, we report that circular PVT1 (circPVT1) was upregulated in the left ventricle of 31 ischemic heart failure patients compared to 11 non-ischemic controls. RNA sequencing analysis following circPVT1 knockdown in immortalized human cardiomyocytes identified differentially expressed genes, mainly involved in fibrosis. Notably, in human cardiac fibroblasts, circPVT1 expression significantly increased after TGF-β1 treatment and circPVT1 silencing attenuated the levels of pro-fibrotic markers induced by TGF-β1. RNA pull-down assays validated the interaction between circPVT1 and two fibrosis-related miRNAs, miR-30a-5p and miR-125b-5p. The levels of these miRNAs were not altered upon circPVT1 knockdown. However, the expression of their mRNA targets was deregulated upon circPVT1 silencing, suggesting that circPVT1 modulates miRNA cellular bioavailability. Accordingly, inhibition of either miR-30a-5p or miR-125b-5p restored the expression of TGF-β1-induced pro-fibrotic markers following circPVT1 silencing, indicating that both miR-30a-5p and miR-125b-5p act as downstream effectors of circPVT1 in cardiac fibroblast activation. In conclusion, these findings highlight a pro-fibrotic role for circPVT1, which can regulate cardiac fibroblast activation interacting with the anti-fibrotic miR-30a-5p and miR-125b-5p. The modulation of circPVT1 expression may represent a potential strategy to reduce cardiac fibrosis and remodeling.
Collapse
Affiliation(s)
- Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | | | - Anna Sofia Tascini
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Spyros Tastsoglou
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Germana Zaccagnini
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Department of Mechanical Circulatory Support and Transplant, National Institute of Cardiology, Warsaw, Poland
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania.
| |
Collapse
|
4
|
Jiang H, Zhou Y, Zhang W, Li H, Ma W, Ji X, Zhou C. Molecular mechanisms of endothelial-mesenchymal transition and its pathophysiological feature in cerebrovascular disease. Cell Biosci 2025; 15:49. [PMID: 40253404 PMCID: PMC12008988 DOI: 10.1186/s13578-025-01393-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
The phenomenon of endothelial-mesenchymal transition (EndMT), a distinct subtype of epithelial-mesenchymal transition (EMT), has garnered significant attention from scholars. EndMT refers to the process whereby endothelial cells (ECs) transform into mesenchymal cells in response to various stimuli, resulting in the loss of their original characteristics. This process has diverse implications in both physiological and pathological states. Under physiological conditions, EndMT plays a crucial role in the development of the cardiovascular system. Conversely, under pathological conditions, EndMT has been identified as a pivotal factor in the development of cardiovascular diseases. Nonetheless, a comprehensive overview of EndMT in cerebrovascular disease is currently lacking. Here, we discuss the heterogeneity of EndMT occurrence and the regulatory factors involved in its development and analyze the feasibility of EndMT as a therapeutic target, aiming to provide a solid theoretical foundation and evidence to address diseases caused by pathological EndMT.
Collapse
Affiliation(s)
- Huimin Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China
| | - Weiyue Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Hui Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
5
|
Peng Y, Mei S, Qi X, Tang R, Yang W, Feng J, Zhou Y, Huang X, Qian G, Xing S, Gao Y, Xu Q, He Z. PGC-1α mediates migrasome secretion accelerating macrophage-myofibroblast transition and contributing to sepsis-associated pulmonary fibrosis. Exp Mol Med 2025:10.1038/s12276-025-01426-z. [PMID: 40164683 DOI: 10.1038/s12276-025-01426-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/29/2024] [Accepted: 01/01/2025] [Indexed: 04/02/2025] Open
Abstract
Sepsis-associated pulmonary fibrosis (SAPF) is a critical pathological stage in the progression of sepsis-induced acute respiratory distress syndrome. While the aggregation and activation of lung fibroblasts are central to the initiation of pulmonary fibrosis, the macrophage-myofibroblast transition (MMT) has recently been identified as a novel source of fibroblasts in this context. However, the mechanisms driving MMT remain inadequately understood. Given the emerging role of migrasomes (novel extracellular vesicles mediating intercellular communication), we investigated their involvement in pulmonary fibrosis. Here we utilized a lipopolysaccharide-induced SAPF mouse model and an in vitro co-culture system of fibroblasts and macrophages to observe the MMT process during SAPF. We found that lipopolysaccharide exposure suppresses PGC-1α expression in lung fibroblasts, resulting in mitochondrial dysfunction and the accumulation of cytosolic mitochondrial DNA (mtDNA). This dysfunction promotes the secretion of mtDNA-containing migrasomes, which, in turn, initiate the MMT process and contribute to fibrosis progression. Notably, the activation of PGC-1α mitigates mitochondrial dysfunction, reduces mtDNA-migrasome release, inhibits MMT and alleviates SAPF. In conclusion, our study identifies the suppression of PGC-1α in lung fibroblasts and the subsequent release of mtDNA migrasomes as a novel mechanism driving MMT in SAPF. These findings suggest that targeting the crosstalk between fibroblasts and immune cells mediated by migrasomes could represent a promising therapeutic strategy for SAPF.
Collapse
Affiliation(s)
- Yawen Peng
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shuya Mei
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Xiaohui Qi
- Department of Cardiovascular Surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ri Tang
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Wenyu Yang
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Jinhua Feng
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Yang Zhou
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Xi Huang
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Guojun Qian
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shunpeng Xing
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Gao
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qiaoyi Xu
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| | - Zhengyu He
- Department of Critical Care Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China.
| |
Collapse
|
6
|
Bei Y, Shen ZX, Lin HR, Wei TF, Wang YH, Su ZT, Dai YJ, Wang YH, Huang LL, Zhu T, Hu W, Ye J, Wu GX, Dai HB. Endothelial histone deacetylase 9 promotes diabetic retinopathy in mice by regulating endothelial-mesenchymal transition. Acta Pharmacol Sin 2025:10.1038/s41401-025-01523-9. [PMID: 40164754 DOI: 10.1038/s41401-025-01523-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/23/2025] [Indexed: 04/02/2025]
Abstract
Diabetic retinopathy (DR) is a common and specific microvascular complication of diabetes and the leading cause of blindness in working-age adults. Endothelial-mesenchymal transition (EndoMT) underlies various chronic vascular diseases, while histone deacetylase 9 (HDAC9) is involved in the pathological process of cardiovascular diseases, cerebrovascular diseases, autoimmune diseases, and breast cancer. Recent evidence has shown that HDAC9 promotes EndoMT, thereby affecting the progression of atherosclerotic disease. In this study, we investigated the critical role of HDAC9 in DR and the underlying mechanism. DR model was established in mice by injecting streptozotocin (STZ, 50 mg/kg) for 5 consecutive days. Blood glucose was monitored regularly and DR experiments were performed 12 weeks after modeling. We showed that the expression levels of HDAC9 were significantly elevated in the vitreous fluid of diabetic patients and the retinal endothelial cells of DR model mice. Knockdown of endothelial HDAC9 reduced EndoMT and alleviated DR pathology in vivo, whereas overexpression of HDAC9 exacerbated EndoMT in DR model mice. To elucidate the downstream target genes of HDAC9 implicated in DR, we conducted integrated ChIP-seq and RNA-seq analysis of the retina in STZ-induced retinopathy and established that HDAC9 was involved in the transcriptional regulation of annexin A2 (ANXA2). We demonstrated that HDAC9 was bound to the promoter region of ANXA2, leading to the downregulation of ANXA2 expression in high glucose-treated human retinal microvascular endothelial cells and STZ-induced DR model mice. Overexpression of ANXA2 significantly reduced the EndoMT process in STZ-induced DR model mice. Collectively, our results demonstrate that HDAC9 promotes EndoMT by regulating ANXA2 transcription, thereby disrupting vascular homeostasis during DR. This study sheds light on the roles of HDAC9 and ANXA2 in DR pathology and provides a theoretical foundation for the potential therapeutic strategies to target DR.
Collapse
Affiliation(s)
- Yun Bei
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Department of Pharmacy, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, 313000, China
| | - Ze-Xu Shen
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Hao-Ran Lin
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tao-Feng Wei
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yi-Hao Wang
- The High School Affiliated of Renmin University of China, Beijing, 100080, China
| | - Zhi-Tao Su
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yun-Jian Dai
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yan-Hong Wang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Ling-Ling Huang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tao Zhu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Wei Hu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Gong-Xiong Wu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Department of Medicine, Laboratory for Translational Research, Harvard Medical School, Cambridge, MA, USA.
| | - Hai-Bin Dai
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
7
|
Carollo C, Sorce A, Cirafici E, Mulè G, Caimi G. Sirtuins and Resveratrol in Cardiorenal Diseases: A Narrative Review of Mechanisms and Therapeutic Potential. Nutrients 2025; 17:1212. [PMID: 40218970 PMCID: PMC11990745 DOI: 10.3390/nu17071212] [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: 03/14/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
Aging is a very complex process, and it has been linked with Sirtuins. Sirtuin enzymes are a family of deacetylases that are related to caloric restriction and aging by modulating energy metabolism, genomic stability, and stress resistance. Up to now, seven sirtuins have been recognized. This narrative review aimed to analyze the literature produced between January 2005 and March 2025 to evaluate the role of sirtuins in chronic kidney disease and, as heart and kidney diseases are strictly interrelated, to explore their role in heart diseases and cardio-renal cross-talk. A reciprocal relationship between CKD and aging seems to exist since CKD may contribute to premature biological aging of different organ systems. SIRTs are involved in the pathophysiology of renal diseases; their activation can delay the progression of several renal diseases. Notably, an increasing number of studies linked SIRTs with different CVDs. SIRTs affect the production of mitochondrial reactive oxygen species (ROS) by modulating mitochondrial function. The imbalance of SIRT levels may increase the vulnerability to CVDs. SIRTs are involved in the pathophysiological mechanisms of HFpEF (heart failure with preserved ejection fraction) through different signaling pathways. Fibrosis is the linkage mechanism between the heart and kidney in the development of cardio-renal diseases. Current studies on sirtuins, resveratrol, and cardiorenal disease highlight their potential therapeutic benefits in regulating blood pressure, kidney function, lipid profiles, and inflammation, making them a promising area of investigation for improving cardiovascular and renal health outcomes. However, significant gaps remain. The limited availability of highly selective and potent sirtuin modulators hampers their clinical translation, as most existing compounds exhibit poor bioavailability and suboptimal pharmacokinetic properties.
Collapse
Affiliation(s)
- Caterina Carollo
- Department of Health Promotion, Mother and Child Care, Internal and Specialistic Medicine, University of Palermo, 90127 Palermo, Italy (E.C.); (G.M.)
| | | | | | | | | |
Collapse
|
8
|
Punde A, Rayrikar A, Maity S, Patra C. Extracellular matrix in cardiac morphogenesis, fibrosis, and regeneration. Cells Dev 2025:204023. [PMID: 40154789 DOI: 10.1016/j.cdev.2025.204023] [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: 10/17/2024] [Revised: 03/14/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in providing structural integrity and regulating cell communication essential for organ development, homeostasis, and regeneration, including hearts. Evidence indicates that disruptions in the spatiotemporal expression or alterations in ECM components lead to cardiac malformations, including a wide range of congenital heart diseases (CHDs). Furthermore, research on injured hearts across various vertebrate species, some of which show effective regeneration while others experience irreversible fibrosis, underscores the significance of ECM molecules in cardiac regeneration. This review presents an overview of heart development and the dynamics of ECM during cardiac morphogenesis, beginning with the formation of the contractile heart tube and advancing to the development of distinct chambers separated by valves to facilitate unidirectional blood flow. Furthermore, we discuss research emphasizing the multifaceted roles of secreted molecules in mediating fibrosis and regeneration following myocardial injury.
Collapse
Affiliation(s)
- Ashwini Punde
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Amey Rayrikar
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Shreya Maity
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India
| | - Chinmoy Patra
- Department of Developmental Biology, Agharkar Research Institute, Pune, Maharashtra, 411004, India.
| |
Collapse
|
9
|
Carlson WD, Bosukonda D, Keck PC, Bey P, Tessier SN, Carlson FR. Cardiac preservation using ex vivo organ perfusion: new therapies for the treatment of heart failure by harnessing the power of growth factors using BMP mimetics like THR-184. Front Cardiovasc Med 2025; 12:1535778. [PMID: 40171539 PMCID: PMC11960666 DOI: 10.3389/fcvm.2025.1535778] [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: 11/27/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
As heart transplantation continues to be the gold standard therapy for end-stage heart failure, the imbalance between the supply of hearts, and the demand for them, continues to get worse. In the US alone, with less than 4,000 hearts suitable for transplant and over 100,000 potential recipients, this therapy is only available to a very few. The use of hearts Donated after Circulatory Death (DCD) and Donation after Brain Death (DBD) using ex vivo machine perfusion (EVMP) is a promising approach that has already increased the availability of suitable organs for heart transplantation. EVMP offers the promise of enabling the expansion of the overall number of heart transplants and lower rates of early graft dysfunction. These are realized through (1) safe extension of the time between procurement and transplantation and (2) ex vivo assessment of preserved hearts. Notably, ex vivo perfusion has facilitated the donation of DCD hearts and improved the success of transplantation. Nevertheless, DCD hearts suffer from serious preharvest ischemia/reperfusion injury (IRI). Despite these developments, only 40% of hearts offered for transplantation can be utilized. These devices do offer an opportunity to evaluate donor hearts for transplantation, resuscitate organs previously deemed unsuitable for transplantation, and provide a platform for the development of novel therapeutics to limit cardiac injury. Bone Morphogenetic Protein (BMP) signaling is a new target which holds the potential for ameliorating myocardial IRI. Recent studies have demonstrated that BMP signaling has a significant role in blocking the deleterious effects of injury to the heart. We have designed novel small peptide BMP mimetics that act via activin receptor-like kinase (ALK3), a type I BMP receptor. They are capable of (1) inhibiting inflammation and apoptosis, (2) blocking/reversing the epithelial-mesenchymal transition (EMT) and fibrosis, and (3) promoting tissue regeneration. In this review, we explore the promise that novel therapeutics, including these BMP mimetics, offer for the protection of hearts against myocardial injury during ex vivo transportation for cardiac transplantation. This protection represents a significant advance and a promising ex vivo therapeutic approach to expanding the donor pool by increasing the number of transplantable hearts.
Collapse
Affiliation(s)
- William D. Carlson
- Division of Cardiology, Mass General Hospital/Harvard, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Therapeutics by Design, Weston, MA, United States
| | - Dattatreyamurty Bosukonda
- Division of Cardiology, Mass General Hospital/Harvard, Boston, MA, United States
- Therapeutics by Design, Weston, MA, United States
| | | | - Philippe Bey
- Therapeutics by Design, Weston, MA, United States
| | - Shannon N. Tessier
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Children’s Hospital, Boston, MA, United States
| | | |
Collapse
|
10
|
Zhang Z, Yan Z, Yuan T, Zhao X, Wang M, Liu G, Gan L, Qin W. PD-1 inhibition disrupts collagen homeostasis and aggravates cardiac dysfunction through endothelial-fibroblast crosstalk and EndMT. Front Pharmacol 2025; 16:1549487. [PMID: 40166462 PMCID: PMC11955664 DOI: 10.3389/fphar.2025.1549487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Cardiac immune-related adverse events (irAEs) from PD-1-targeting immune check-point inhibitors (ICIs) are an increasing concern due to their high mortality rate. Collagen plays a crucial role in maintaining cardiac structure, elasticity, and signal transduction; however, the effects and mechanisms of PD-1 inhibitor on cardiac collagen remodeling remain poorly understood. Methods C57BL/6 mice were injected with anti-mouse PD-1 antibody to create a PD-1 inhibitor-treated model. Cardiac function was measured by echocardiography, and collagen distribution was analyzed with Masson's trichrome staining and Sirius Red staining. Single-nucleus RNA sequencing was performed to examine the effects of PD-1 inhibition on gene expression in cardiac fibroblasts (CFs) and endothelial cells (ECs). EC-CF crosstalk was assessed using co-culture experiments and ELISA. ChIP assay was performed to analyze the regulation of TCF12 on TGF-β1 promoter. Western blot, qRT-PCR, and immunofluorescence staining were used to detect the expression of TCF12, TGF-β1, and endothelial-to-mesenchymal transition (EndMT) markers. Reactive oxygen species (ROS) levels were evaluated by DHE staining, MDA content, and SOD activity assays. Results We report a newly discovered cardiotoxic effect of PD-1 inhibitor, which causes aberrant collagen distribution in the heart, marked by a decrease in interstitial collagen and an increase in perivascular collagen deposition. Mechanistically, PD-1 inhibitor does not directly affect CFs but instead impact them through EC-CF crosstalk. PD-1 inhibitor reduces TGF-β1 secretion in ECs by downregulating TCF12, which we identify as a transcriptional promoter of TGF-β1. This subsequently decreases CF activity, leading to reduced interstitial collagen deposition. Additionally, PD-1 inhibitor induces EndMT, increasing perivascular collagen deposition. The endothelial dysfunction induced by PD-1 inhibitor results from ROS accumulation in ECs. Inhibiting ROS with N-acetylcysteine (NAC) preserves normal collagen distribution and cardiac function in PD-1 inhibitor-treated mice by reversing TCF12 downregulation and EndMT in ECs. Conclusion Our results suggest that PD-1 inhibitor causes ROS accumulation in cardiac ECs, leading to imbalanced collagen distribution (decrease in interstitial collagen and increase in perivascular collagen) in the heart by modulating TCF12/TGF-β1-mediated EC-CF crosstalk and EndMT. NAC supplementation could be an effective clinical strategy to mitigate PD-1 inhibitor-induced imbalanced collagen distribution and cardiac dysfunction.
Collapse
Affiliation(s)
- Zejin Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
| | - Zhenzhen Yan
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong University of Traditional Chinese medicine, Jinan, Shandong, China
| | - Tao Yuan
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong First Medical University, Jinan, Shandong, China
| | - Xiaona Zhao
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong Second Medical University, Weifang, Shandong, China
| | - Minghui Wang
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong University of Traditional Chinese medicine, Jinan, Shandong, China
| | - Guoqing Liu
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- School of Pharmacy, Shandong University of Traditional Chinese medicine, Jinan, Shandong, China
| | - Lijun Gan
- Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment), Affiliated Hospital of Jining Medical University, Jining, Shandong, China
| | - Wei Qin
- School of Pharmacy, Jining Medical University, Rizhao, Shandong, China
- Department of Cardiology (Shandong Provincial Key Laboratory for Cardiovascular Disease Diagnosis and Treatment), Affiliated Hospital of Jining Medical University, Jining, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
11
|
Wei Y, Li X, Sha Z, Liu J, Wu G, Zhou T, Lin C, Xie Y, Bao Y, Luo Q, Ling T, Pan W, Xie Y, Zhang N, Jin Q, Wu L. Exosomal Prolactin-Induced Protein Inhibits the Activation of cGMP/PKG Pathway Mediated by ATP2B2 to Promote Myocardial Fibrosis in Atrial Fibrillation. Antioxid Redox Signal 2025. [PMID: 40094760 DOI: 10.1089/ars.2024.0723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Aims: Myocardial fibrosis is an important medium for atrial fibrillation (AF). Exosomes have been demonstrated to affect the development of AF. This study explored the molecular mechanism of exosomes from patients with AF (AF-exo) mediating myocardial fibrosis and thus affecting the development of AF. Results: Prolactin-induced protein (PIP) is highly expressed in AF-exo. AF-exo promoted the proliferation and activation of cardiac fibroblasts (CFs) as well as the migration and endothelial-to-mesenchymal transition (EndMT) of human umbilical vein endothelial cells (HUVECs). However, the effect of AF-exo on CFs and HUVECs was mitigated by PIP-specific short hairpin RNA (shPIP). Adeno-associated virus (AAV)-shPIP reduced the incidence and duration of AF in rats, and improved myocardial fibrosis and collagen deposition. ATPase plasma membrane Ca2+ transporting 2 (ATP2B2) overexpression or inhibition reverses the role of PIP or shPIP in CFs, HUVECs, and AF rats. Activation of the cyclic guanosine monophosphate/protein kinase G (cGMP/PKG) pathway is beneficial to alleviate myocardial fibrosis, but this effect is mitigated by shATP2B2. Innovation: Our investigation substantiates the pivotal role of the PIP/ATP2B2 axis in both HUVEC myocardial fibrosis and EndMT progression. Our findings suggest that AF-exo can suppress the activation of the cGMP/PKG pathway mediated by ATP2B2 through exosomal PIP, thus promoting myocardial fibrosis, indicating potential targets for novel antifibrotic drug development targeting either PIP or ATP2B2. Conclusion: Exosomal PIP can inhibit the activation of cGMP/PKG pathway mediated by ATP2B2, thus promoting the development of AF. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Yue Wei
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Li
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zimo Sha
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingmeng Liu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanhua Wu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Taojie Zhou
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changjian Lin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Bao
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingzhi Luo
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyou Ling
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqi Pan
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yucai Xie
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Jin
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liqun Wu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Li Z, Yin YJ, Wei YR, Liu Y, Han NX, Wang XQ, Hao YJ, Wang YF, Hou YL, Jia ZH. Protective role of Tongxinluo in mitigating myocardial fibrosis in mice with acute myocardial infarction via neuregulin-1 upregulation and Inhibition of endothelium-interstitial transition. J Mol Histol 2025; 56:103. [PMID: 40063284 PMCID: PMC11893718 DOI: 10.1007/s10735-025-10378-7] [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: 11/06/2024] [Accepted: 02/17/2025] [Indexed: 03/14/2025]
Abstract
Acute myocardial infarction (AMI) is a leading cause of heart failure, often accompanied by myocardial fibrosis (MF), characterized by excessive extracellular matrix accumulation. Endothelial-to-mesenchymal transition (EndMT) plays a key role in MF progression post-AMI. Neuregulin-1 (NRG-1), a growth factor with cardioprotective properties, has emerged as a potential therapeutic target. Tongxinluo (TXL), a traditional Chinese medicine, mitigates MF by upregulating NRG-1. This study elucidates the mechanisms underlying the protective effects of NRG-1 and TXL against MF following AMI. Left anterior descending artery ligation established a model for mice with AMI. Adeno-associated virus was used to modulate NRG-1 expression in the myocardium. Echocardiography assessed cardiac function, and histological staining was used to evaluate MF. Expression levels of markers for myofibroblasts (α-SMA, FSP-1) and endothelial cells (CD31, VE-cadherin) were analysed to investigate EndMT. The involvement of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) signalling pathway in NRG-1's protective mechanism was validated using biochemical methods. Tongxinluo was administered to mice with AMI via gavage for 4 weeks, and its effects on cardiac function, MF and EndMT were assessed. Overexpression of NRG-1 in mice with AMI ameliorated cardiac dysfunction and reduced interstitial and perivascular fibrosis, whereas NRG-1 deficiency exacerbated these effects. NRG-1 protected against EndMT, as evidenced by changes in myofibroblast and endothelial cell markers. The PI3K/AKT signalling pathway was involved in NRG-1's protective mechanism against MF. The administration of TXL to mice with AMI improved cardiac function and reduced MF by activating NRG-1. Furthermore, TXL inhibited EndMT post-AMI through the NRG-1/PI3K/AKT pathway. NRG-1 and TXL protect against MF post-AMI by mitigating EndMT through the PI3K/AKT pathway. These findings suggest that targeting NRG-1 or using TXL may be promising therapeutic strategies for MF following AMI.
Collapse
Affiliation(s)
- Zhen Li
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Yu-Jie Yin
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Ya-Ru Wei
- Hebei Yiling Hospital, Shijiazhuang, 050091, China
| | - Yi Liu
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Ning-Xin Han
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Xiao-Qi Wang
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China
| | - Yuan-Jie Hao
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China
| | - Ya-Fen Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China
| | - Yun-Long Hou
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China.
| | - Zhen-Hua Jia
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei, 050017, China.
- Hebei Academy of Integrated Traditional Chinese and Western Medicine, Shijiazhuang, 050035, China.
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang, 050035, China.
- Hebei Yiling Hospital, Shijiazhuang, 050091, China.
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, 050090, China.
| |
Collapse
|
13
|
Yang YF, Holden P, Sun YY, Faralli JA, Peters DM, Keller KE. Fibrosis-Related Gene and Protein Expression in Normal and Glaucomatous Trabecular Meshwork Cells. Invest Ophthalmol Vis Sci 2025; 66:48. [PMID: 40126508 PMCID: PMC11951066 DOI: 10.1167/iovs.66.3.48] [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: 12/19/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025] Open
Abstract
Purpose Glaucomatous trabecular meshwork (GTM) tissue is characterized by excess fibrotic-like extracellular matrices, which negatively impacts aqueous humor outflow. Endothelial-to-mesenchymal transition (EndMT) is the process by which tissues develop fibrosis. In this study, we investigated fibrotic-related gene and protein profiles of non-glaucomatous trabecular meshwork (NTM) and GTM cells. Methods Primary cells were cultured from NTM (n = 6) and GTM (n = 5) age-matched cadaver eyes. RNA was harvested and mRNA profiling of 750 genes was performed using the human fibrosis panel (NanoString). Quantitative PCR (qPCR), Western blotting, and immunofluorescence microscopy were performed. A matrix metalloproteinase (MMP) fluorogenic assay was used to quantitate enzyme activity. Results Classic EndMT biomarkers, α-SMA, SNAI2, TWIST1, TWIST2, and VIM, were upregulated in GTM cells, whereas increased phosphorylated SMAD2-3 indicated increased TGFβ signaling. GTM cells had increased deposition of FN-EDA fibronectin fibrils, but reduced amounts of FN-EDB fibrils, and altered immunostaining of active α5β1 and αvβ3 integrins. NanoString analysis showed that 2 genes were upregulated and 28 genes were downregulated in GTM cells compared with NTM cells. Western immunoblotting confirmed increased protein levels of N-cadherin and decreased MMP2, CHI3L1, COL6A3, and SERPINF1 proteins in GTM cells. Whereas MMP2 gene and protein levels were reduced, there was increased MMP activity. Conclusions Increased expression of α-SMA, FN-EDA, N-cadherin, SNAI2, TWISTs, VIM, TGFβ signaling, and MMP activity are consistent with GTM cells acquiring an EndMT phenotype. In combination with tissue studies, cultured GTM cells are a useful in vitro model for studying the fibrotic process in glaucoma.
Collapse
Affiliation(s)
- Yong-Feng Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Paul Holden
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Ying Ying Sun
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Jennifer A. Faralli
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States
| | - Donna M. Peters
- Departments of Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States
- Ophthalmology and Visual Sciences, University of Wisconsin, Madison, Wisconsin, United States
| | - Kate E. Keller
- Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
14
|
Xu H, Mao X, Wang Y, Zhu C, Liang B, Zhao Y, Zhou M, Ye L, Hong M, Shao H, Wang Y, Li H, Qi Y, Yang Y, Chen L, Guan Y, Zhang X. Targeting the E Prostanoid Receptor EP4 Mitigates Cardiac Fibrosis Induced by β-Adrenergic Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413324. [PMID: 39921269 PMCID: PMC11948031 DOI: 10.1002/advs.202413324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/10/2025] [Indexed: 02/10/2025]
Abstract
Sustained β-adrenergic activation induces cardiac fibrosis characterized by excessive deposition of extracellular matrix (ECM). Prostaglandin E2 (PGE2) receptor EP4 is essential for cardiovascular homeostasis. This study aims to investigate the roles of cardiomyocyte (CM) and cardiac fibroblast (CF) EP4 in isoproterenol (ISO)-induced cardiac fibrosis. By crossing the EP4f/f mice with α-MyHC-Cre or S100A4-Cre mice, this work obtains the CM-EP4 knockout (EP4f/f-α-MyHCCre+) or CF-EP4 knockout (EP4f/f-S100A4Cre+) mice. The mice of both genders are subcutaneously injected with ISO (5 mg kg-1 day-1) for 7 days. Compared to the control mice, both EP4f/f-α-MyHCCre+ and EP4f/f-S100A4Cre+ mice show a significant improvement in cardiac diastolic function and fibrosis as assessed by echocardiography and histological staining, respectively. In the CMs, inhibition of EP4 suppresses ISO-induced TGF-β1 expression via blocking the cAMP/PKA pathway. In the CFs, inhibition of EP4 reversed TGF-β1-triggers production of ECM via preventing the formation of the TGF-β1/TGF-β receptor complex and blocks CF proliferation via suppressing the ERK1/2 pathway. Furthermore, double knockout of the CM- and CF-EP4 or administration of EP4 antagonist, grapiprant, markedly improves ISO-induced cardiac diastolic dysfunction and fibrosis. Collectively, this study demonstrates that both CM-EP4 and CF-EP4 contribute to β-adrenergic activation-induced cardiac fibrosis. Targeting EP4 may offer a novel therapeutic approach for cardiac fibrosis.
Collapse
Affiliation(s)
- Hu Xu
- Wuhu HospitalEast China Normal UniversityShanghai200241China
- Health Science CenterEast China Normal UniversityShanghai200241China
| | - Xiuhui Mao
- Health Science CenterEast China Normal UniversityShanghai200241China
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Yali Wang
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Chunhua Zhu
- Health Science CenterEast China Normal UniversityShanghai200241China
| | - Bo Liang
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Yihang Zhao
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Mengfei Zhou
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Lan Ye
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Mengting Hong
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Huishu Shao
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Yashuo Wang
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Haonan Li
- MOE Key Laboratory of Bio‐Intelligent ManufacturingSchool of BioengineeringDalian University of TechnologyDalian116024China
| | - Yinghui Qi
- Department of NephrologyPudong New District Punan HospitalShanghai200125China
| | - Yongliang Yang
- MOE Key Laboratory of Bio‐Intelligent ManufacturingSchool of BioengineeringDalian University of TechnologyDalian116024China
| | - Lihong Chen
- Health Science CenterEast China Normal UniversityShanghai200241China
| | - Youfei Guan
- Health Science CenterEast China Normal UniversityShanghai200241China
- Advanced Institute for Medical SciencesDalian Medical UniversityDalian116044China
| | - Xiaoyan Zhang
- Wuhu HospitalEast China Normal UniversityShanghai200241China
- Health Science CenterEast China Normal UniversityShanghai200241China
| |
Collapse
|
15
|
Erdogan BR, Arioglu-Inan E. SGLT2 inhibitors: how do they affect the cardiac cells. Mol Cell Biochem 2025; 480:1359-1379. [PMID: 39160356 DOI: 10.1007/s11010-024-05084-z] [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: 10/27/2023] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
The first sodium-glucose cotransporter-2 inhibitor (SGLT2I), canagliflozin, was approved by the U.S. Food and Drug Administration for the treatment of type 2 diabetes in 2013. Since then, other members of this drug class (such as dapagliflozin, empagliflozin, and ertugliflozin) have become widely used. Unlike classical antidiabetic agents, these drugs do not interfere with insulin secretion or action, but instead promote renal glucose excretion. Since their approval, many preclinical and clinical studies have been conducted to investigate the diverse effects of SGLT2Is. While originally introduced as antidiabetic agents, the SGLT2Is are now recognized as pillars in the treatment of heart failure and chronic kidney disease, in patients with or without diabetes. The beneficial cardiac effects of this class have been attributed to several mechanisms. Among these, SGLT2Is inhibit fibrosis, hypertrophy, apoptosis, inflammation, and oxidative stress. They regulate mitochondrial function and ion transport, and stimulate autophagy through several underlying mechanisms. This review details the potential effects of SGLT2Is on cardiac cells.
Collapse
Affiliation(s)
| | - Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Emniyet District, Dogol Street, No:4, 06560, Yenimahalle, Ankara, Turkey.
| |
Collapse
|
16
|
Timmer LT, den Hertog E, Versteeg D, Post H, Verdonschot JAJ, Monshouwer-Kloots J, Kyriakopoulou E, Perini I, Koopmans T, van der Kraak P, Zentilin L, Heymans SRB, Vink A, Giacca M, Heck AJR, van Rooij E. Cardiomyocyte SORBS2 expression increases in heart failure and regulates integrin interactions and extracellular matrix composition. Cardiovasc Res 2025:cvaf021. [PMID: 39957251 DOI: 10.1093/cvr/cvaf021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/26/2024] [Accepted: 12/05/2024] [Indexed: 02/18/2025] Open
Abstract
AIMS In this study, we aimed to uncover genes associated with stressed cardiomyocytes by combining single-cell transcriptomic datasets from failing cardiac tissue from both humans and mice. METHODS AND RESULTS Our bioinformatic analysis identified SORBS2 as conserved NPPA correlated gene. Using mouse models and cardiac tissue from human heart failure patients, we demonstrated that SORBS2 expression is consistently increased during pathological remodeling, correlates to disease severity and is regulated by GATA4. By affinity-purification mass-spectrometry, we showed SORBS2 to interact with the integrin-cytoskeleton connections. Cardiomyocyte-specific genetic loss of Sorbs2 in adult mice changed integrin interactions, indicated by the increased expression of several integrins and altered extracellular matrix components connecting to these integrins, leading to an exacerbated fibrotic response during pathological remodeling. CONCLUSIONS Sorbs2 is a cardiomyocyte-enriched gene that is increased during progression to heart failure in a GATA4-dependent manner and correlates to phenotypical hallmarks of cardiac failure.Our data indicate SORBS2 to function as a crucial regulator of integrin interactions and cardiac fibrosis.
Collapse
Affiliation(s)
- Louk T Timmer
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Elvira den Hertog
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Job A J Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart)
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Eirini Kyriakopoulou
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ilaria Perini
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Petra van der Kraak
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lorena Zentilin
- Molecular Medicine Laboratory, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Stephane R B Heymans
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium
| | - Aryan Vink
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mauro Giacca
- Molecular Medicine Laboratory, International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- School of Cardiovascular and Metabolic Medicine & Sciences and British Heart Foundation Centre of Research Excellence, King's College London, London, UK
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics Group, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
17
|
Li H, Jia Y, Chen Z, Yang L, Ni L, Cao Y, Fan R, Yuan Z, Zhu K, Zhang Z, Zuo L, Wu P, Gao Y, Lin Y. Bioinformatics analysis of coronary microvascular dysfunction in rats based on single-cell RNA sequencing. Sci Rep 2025; 15:5050. [PMID: 39934189 DOI: 10.1038/s41598-025-85318-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/01/2025] [Indexed: 02/13/2025] Open
Abstract
Coronary microvascular dysfunction serves as one of the etiological factors for ischemic heart disease and represents a novel therapeutic direction for coronary artery diseases; however, the research on its pathogenesis remains inconsistent. This study aims to explore the single-cell gene expression profiles in rats with coronary microvascular dysfunction using single-cell RNA sequencing, with a particular focus on the in-depth analysis of endothelial cell gene expression characteristics. By establishing a rat model of coronary microvascular dysfunction, we collected cardiac apical tissue to prepare single-cell suspensions and further analyzed them using bioinformatics methods. From a total of 55,419 cells, we identified 28 distinct cell clusters, with endothelial cells and fibroblasts being the predominant cell types. Compared to the NC group, the proportion of endothelial cells in the CMD group was significantly reduced, while the number of fibroblasts was significantly increased. Through further analysis of the endothelial cells, we classified them into normal phenotype endothelial cells, mesenchymal phenotype endothelial cells, proliferative phenotype endothelial cells, and lymphatic endothelial cells, with mesenchymal and proliferative endothelial cells originating from normal phenotype endothelial cells. Additionally, the CMD group exhibited an increase in immune cells, enhanced inflammatory response, and increased oxidative stress. These findings may provide novel potential therapeutic targets for the treatment of Coronary microvascular dysfunction.
Collapse
Affiliation(s)
- Hao Li
- LinFen Central Hospital, Department of Cardiology, LinFen, 041000, China
| | - Yiding Jia
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Zelin Chen
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Luqun Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Lin Ni
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Yuchen Cao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Rong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Zitong Yuan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Zhijun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China
| | - Lin Zuo
- Shanxi Medical UniversityLaboratory of Cell Physiology, Jinzhong, 030600, China
| | - Ping Wu
- First Hospital of Shanxi Medical University, Department of Cardiology, Taiyuan, 030012, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China.
| | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Department of Cardiology, 99 Longcheng Street, Xiaodian District, Taiyuan, 030032, Shanxi, China.
| |
Collapse
|
18
|
Figueroa-Juárez E. Tracing the roots of cardiac fibrosis: role of endothelial-to-mesenchymal transition. Nat Rev Cardiol 2025:10.1038/s41569-025-01134-1. [PMID: 39920247 DOI: 10.1038/s41569-025-01134-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
|
19
|
Li Y, Xing Y, Liu N, Liu B, Wang Z. SOX9: a key transcriptional regulator in organ fibrosis. Front Pharmacol 2025; 16:1507282. [PMID: 39974732 PMCID: PMC11835943 DOI: 10.3389/fphar.2025.1507282] [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: 10/07/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
The SOX9 gene locus is not only extensive but also intricate, and it could promote fibrosis in different organs or tissues, including cardiac fibrosis, liver fibrosis, kidney fibrosis, pulmonary fibrosis, as well as other organ fibrosis. Many disorders are associated with the process of fibrosis; moreover, fibrosis is a common symptom of chronic inflammatory diseases, characterized by the accumulation of excessive components in the extracellular matrix through different signaling pathways. The advanced stage of the fibrotic process leads to organ dysfunction and, ultimately, death. In this review, we first give an overview of the original structure and functions of SOX9. Second, we will discuss the role of SOX9 in fibrosis in various organs or tissues. Third, we describe and reveal the possibility of SOX9 as an antifibrotic treatment target. Finally, we will focus on the application of novel technologies for SOX9 and the subsequent investigation of fibrosis.
Collapse
Affiliation(s)
| | | | | | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Zhihui Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Diaz-Gil D, Silva-Gomez N, Morton SU, Seidman JG, Seidman CE, Zurakowski D, Staffa SJ, Marx GR, Emani SM, Del Nido PJ, Friehs I. Predictive modeling of endocardial fibroelastosis recurrence in patients with congenital heart disease. J Thorac Cardiovasc Surg 2025; 169:366-374. [PMID: 39208926 DOI: 10.1016/j.jtcvs.2024.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Endocardial fibroelastosis (EFE) is a major effector in the maldevelopment of the heart in patients with congenital heart disease. Despite successful surgical removal, EFE can redevelop, but the underlying cause of EFE recurrence remains unknown. This study aimed to identify hemodynamic predictors and genetic links to epithelial/endothelial-to-mesenchymal transition (EMT/EndMT) alterations for preoperative risk assessment. METHODS We assessed the impact of preoperative hemodynamic parameters on EFE recurrence in a cohort of 92 patients with congenital heart disease who underwent left ventricular (LV) EFE resection between January 2010 and March 2021. Additionally, whole-exome sequencing in 18 patients was used to identify rare variants (minor allele frequency <10-5) in high-expression heart (HHE) genes related to cardiac EMT/EndMT and congenital heart disease. RESULTS EFE recurred in 55.4% of patients, within a median of 2.2 years postsurgery. Multivariable analysis revealed specific hemodynamic parameters (mitral valve inflow and area, LV filling pressure, and aortic valve gradient and diameter) as predictors, forming a predictive model with an area under the receiver operating characteristic curve of 0.782. Furthermore, 89% of the patients exhibited damaging variants in HHE genes, with 38% linked to cardiac EMT/EndMT Gene Ontology processes and 22% associated with known congenital heart disease genes. Notably, HHE genes associated with cardiac EMT/EndMT were significantly associated with faster EFE recurrence in a multivariate analysis (hazard ratio, 3.56; 95% confidence interval, 1.24-10.17; P = .018). CONCLUSIONS These findings established a predictive scoring system using preoperative hemodynamic parameters for EFE recurrence risk assessment. Alterations in HHE genes, particularly those linked to cardiac EMT/EndMT, exacerbate the risk of recurrence.
Collapse
Affiliation(s)
- Daniel Diaz-Gil
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Department of Pediatrics, Boston University Chobanian & Avedisian School of Medicine, Boston, Mass; Department of Pediatric Heart Medicine and Adults with Congenital Heart Disease, University Heart and Vascular Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Sarah U Morton
- Department of Pediatrics, Boston Children's Hospital, Boston, Mass; Department of Pediatrics, Harvard Medical School, Boston, Mass; Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital, Boston, Mass
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, Mass; Cardiovascular Division, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass; Howard Hughes Medical Institute, Chevy Chase, Md
| | - David Zurakowski
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston, Mass; Department of Anesthesia, Harvard Medical School, Boston, Mass
| | - Steven J Staffa
- Department of Anesthesiology, Perioperative and Pain Medicine, Boston Children's Hospital, Boston, Mass
| | - Gerald R Marx
- Department of Pediatrics, Harvard Medical School, Boston, Mass; Department of Cardiology, Boston Children's Hospital, Boston, Mass
| | - Sitaram M Emani
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Mass; Department of Surgery, Harvard Medical School, Boston, Mass
| | - Pedro J Del Nido
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Mass; Department of Surgery, Harvard Medical School, Boston, Mass
| | - Ingeborg Friehs
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, Mass; Department of Surgery, Harvard Medical School, Boston, Mass.
| |
Collapse
|
21
|
Zhu L, Liu Y, Wang K, Wang N. Regulated cell death in acute myocardial infarction: Molecular mechanisms and therapeutic implications. Ageing Res Rev 2025; 104:102629. [PMID: 39644925 DOI: 10.1016/j.arr.2024.102629] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Acute myocardial infarction (AMI), primarily caused by coronary atherosclerosis, initiates a series of events that culminate in the obstruction of coronary arteries, resulting in severe myocardial ischemia and hypoxia. The subsequent myocardial ischemia/reperfusion (I/R) injury further aggravates cardiac damage, leading to a decline in heart function and the risk of life-threatening complications. The complex interplay of multiple regulated cell death (RCD) pathways plays a pivotal role in the pathogenesis of AMI. Each RCD pathway is orchestrated by a symphony of molecular regulatory mechanisms, highlighting the dynamic changes and critical roles of key effector molecules. Strategic disruption or inhibition of these molecular targets offers a tantalizing prospect for mitigating or even averting the onset of RCD, thereby limiting the extensive loss of cardiomyocytes and the progression of detrimental myocardial fibrosis. This review systematically summarizes the mechanisms underlying various forms of RCD, provides an in-depth exploration of the pathogenesis of AMI through the lens of RCD, and highlights a range of promising therapeutic targets that hold the potential to revolutionize the management of AMI.
Collapse
Affiliation(s)
- Lili Zhu
- Department of Pathology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yiyang Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China; Key Laboratory of Sepsis Translational Medicine of Hunan, Central South University, Changsha, Hunan, China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan, China.
| |
Collapse
|
22
|
Wang D, Lin M, Wang R, Huang X, Liang Y, Wang X, Chen Y, Gao Y, Guo H, Liang H, Li X. Single-Cell Transcriptomic Reveals the Involvement of Cell-Cell Junctions in the Early Development of Hypertrophic Cardiomyopathy. J Cell Mol Med 2025; 29:e70366. [PMID: 39900554 PMCID: PMC11790354 DOI: 10.1111/jcmm.70366] [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: 07/15/2024] [Revised: 11/06/2024] [Accepted: 01/06/2025] [Indexed: 02/05/2025] Open
Abstract
The relationship between the changes in endothelial cell-cell junctions and microvascular abnormalities in the progression of hypertrophic cardiomyopathy (HCM), as well as their potential as early biomarkers, remains unclear. Here, we analysed single-nucleus RNA-sequencing data from the left ventricles of 44 health donors and HCM patients. First, we observed that endothelial cell-cell junctions were significantly altered in HCM vascular endothelial cells (ECs), including tight junctions, gap junctions and adherens junctions, especially in capillary ECs. The proposed pseudo-timing analysis predicted that endothelial cell-cell junctions abnormalities occurred in the early stages of HCM. Second, we verified that endothelial cell-cell junctions disorders occur at early stages of HCM disease progression in two time-series single-nucleus datasets of mice. The expression of eight cell-cell junction genes showed an initial increase in the early stage, followed by a slight decrease in the middle stage, and a sharp increase in the later stage. Subsequently, cell communication and transcription factor analysis were used to explore the underlying mechanisms. Furthermore, an early HCM prediction model was developed and independently validated using three mRNA datasets comprising 204 health individuals and HCM patients for the eight genes panel, the accuracy was 0.81 [0.63-0.98]. Finally, we validated this panel in HCM tissues. This study demonstrated in humans and mice that eight cell-cell junction genes were significantly elevated in the early stages of HCM and may be potential biomarkers for the early diagnosis of HCM.
Collapse
Affiliation(s)
- Dingchen Wang
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong ProvinceChina
- Department of Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdong ProvinceChina
| | - Miao Lin
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdong ProvinceChina
- Medical Big Data CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdong ProvinceChina
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and ApplicationGuangzhouGuangdong ProvinceChina
| | - Ruobing Wang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdong ProvinceChina
| | - Xiaoran Huang
- Department of Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdong ProvinceChina
| | - Yaowen Liang
- Department of Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdong ProvinceChina
- Shantou University Medical CollegeShantouGuangdong ProvinceChina
| | - Xiran Wang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdong ProvinceChina
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and ApplicationGuangzhouGuangdong ProvinceChina
| | - Yuge Chen
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Yunfei Gao
- Zhuhai Precision Medical CenterZhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan UniversityZhuhaiGuangdong ProvinceChina
- The Biomedical Translational Research InstituteJinan University Faculty of Medical Science, Jinan UniversityGuangzhouGuangdong ProvinceChina
| | - Huiming Guo
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdong ProvinceChina
| | - Huiying Liang
- Guangdong Cardiovascular InstituteGuangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouGuangdong ProvinceChina
- Medical Big Data CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdong ProvinceChina
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and ApplicationGuangzhouGuangdong ProvinceChina
| | - Xin Li
- School of MedicineSouth China University of TechnologyGuangzhouGuangdong ProvinceChina
- Department of Emergency MedicineGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouGuangdong ProvinceChina
| |
Collapse
|
23
|
Huang X, Yu Z, Tian J, Chen T, Wei A, Mei C, Chen S, Li Y. m6A RNA modification pathway: orchestrating fibrotic mechanisms across multiple organs. Brief Funct Genomics 2025; 24:elae051. [PMID: 39756462 PMCID: PMC11735750 DOI: 10.1093/bfgp/elae051] [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: 04/01/2024] [Revised: 09/20/2024] [Accepted: 12/16/2024] [Indexed: 01/07/2025] Open
Abstract
Organ fibrosis, a common consequence of chronic tissue injury, presents a significant health challenge. Recent research has revealed the regulatory role of N6-methyladenosine (m6A) RNA modification in fibrosis of various organs, including the lung, liver, kidney, and heart. In this comprehensive review, we summarize the latest findings on the mechanisms and functions of m6A modification in organ fibrosis. By highlighting the potential of m6A modification as a therapeutic target, our goal is to encourage further research in this emerging field and support advancements in the clinical treatment of organ fibrosis.
Collapse
Affiliation(s)
- Xiangfei Huang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| | - Zilu Yu
- Queen Mary School, Medical College, Nanchang University, 1299 Xuefu Road, Honggutan District, Nanchang 330031, China
| | - Juan Tian
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| | - Tao Chen
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| | - Aiping Wei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| | - Chao Mei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| | - Shibiao Chen
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, 17 Yongwaizheng Street, Donghu District, Nanchang 330006, China
| |
Collapse
|
24
|
Qian C, Dong G, Yang C, Zheng W, Zhong C, Shen Q, Lu Y, Zhao Y. Broadening horizons: molecular mechanisms and disease implications of endothelial-to-mesenchymal transition. Cell Commun Signal 2025; 23:16. [PMID: 39789529 PMCID: PMC11720945 DOI: 10.1186/s12964-025-02028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is defined as an important process of cellular differentiation by which endothelial cells (ECs) are prone to lose their characteristics and transform into mesenchymal cells. During EndMT, reduced expression of endothelial adhesion molecules disrupts intercellular adhesion, triggering cytoskeletal reorganization and mesenchymal transition. Numerous studies have proved that EndMT is a multifaceted biological event driven primarily by cytokines such as TGF-β, TNF-α, and IL-1β, alongside signaling pathways like WNT, Smad, MEK-ERK, and Notch. Nevertheless, the exact roles of EndMT in complicated diseases have not been comprehensively reviewed. In this review, we summarize the predominant molecular regulatory mechanisms and signaling pathways that contribute to the development of EndMT, as well as highlight the contributions of a series of imperative non-coding RNAs in curbing the initiation of EndMT. Furthermore, we discuss the significant impact of EndMT on worsening vasculature-related diseases, including cancer, cardiovascular diseases, atherosclerosis, pulmonary vascular diseases, diabetes-associated fibrotic conditions, and cerebral cavernous malformation, providing the implications that targeting EndMT holds promise as a therapeutic strategy to mitigate disease progression.
Collapse
Affiliation(s)
- Cheng Qian
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanglu Dong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunmei Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weiwei Zheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chongjin Zhong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiuhong Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yang Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
25
|
You N, Liu G, Yu M, Chen W, Fei X, Sun T, Han M, Qin Z, Wei Z, Wang D. Reconceptualizing Endothelial-to-mesenchymal transition in atherosclerosis: Signaling pathways and prospective targeting strategies. J Adv Res 2025:S2090-1232(24)00627-1. [PMID: 39756576 DOI: 10.1016/j.jare.2024.12.049] [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: 09/15/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The modification of endothelial cells (ECs) biological function under pathogenic conditions leads to the expression of mesenchymal stromal cells (MSCs) markers, defined as endothelial-to-mesenchymal transition (EndMT). Invisible in onset and slow in progression, atherosclerosis (AS) is a potential contributor to various atherosclerotic cardiovascular diseases (ASCVD). By triggering AS, EndMT, the "initiator" of AS, induces the progression of ASCVD such as coronary atherosclerotic heart disease (CHD) and ischemic cerebrovascular disease (ICD), with serious clinical complications such as myocardial infarction (MI) and stroke. In-depth research of the pathomechanisms of EndMT and identification of potential targeted therapeutic strategies hold considerable research value for the prevention and treatment of ASCVD-associated with delayed EndMT. Although previous studies have progressively unraveled the complexity of EndMT and its pathogenicity triggered by alterations in vascular microenvironmental factors, systematic descriptions of the most recent pathogenic roles of EndMT in the progression of AS, targeted therapeutic strategies, and their future research directions are scarce. AIM OF REVIEW We aim to provide new researchers with comprehensive knowledge of EndMT in AS. We exhaustively review the latest research advancements in the field and provide a theoretical basis for investigating EndMT, a biological process with sophisticated mechanisms. KEY SCIENTIFIC CONCEPTS OF REVIEW This review summarized that altered hemodynamics with microenvironmental crosstalk consisting of inflammatory responses or glycolysis, oxidative stress, lactate or acetyl-CoA (Ac-CoA), fatty acid oxidation (FAO), intracellular iron overload, and transcription factors, including ELK1 and STAT3, modulate the EndMT and affect AS progression. In addition, we provide new paradigms for the development of promising therapeutic agents against these disease-causing processes and indicate promising directions and challenges that need to be addressed to elucidate the EndMT process.
Collapse
Affiliation(s)
- Nanlin You
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Guohao Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengchen Yu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenbo Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoyao Fei
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Tao Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengtao Han
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhen Qin
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhaosheng Wei
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong 253032, China.
| |
Collapse
|
26
|
Gurgoglione FL, Benatti G, Denegri A, Donelli D, Covani M, De Gregorio M, Dallaglio G, Navacchi R, Niccoli G. Coronary Microvascular Dysfunction: Insights on Prognosis and Future Perspectives. Rev Cardiovasc Med 2025; 26:25757. [PMID: 39867196 PMCID: PMC11760542 DOI: 10.31083/rcm25757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 01/28/2025] Open
Abstract
Coronary microvascular dysfunction (CMD) comprises a wide spectrum of structural and/or functional abnormalities of coronary microcirculation that can lead to myocardial ischemia. Emerging evidence has indicated that CMD is a relevant cause of morbidity and mortality and is associated with a high risk of major adverse cardiovascular events (MACEs) and heart failure with preserved ejection fraction as well as poor quality of life. This review aims to elucidate briefly the pathogenesis and diagnostic modalities of CMD and to shed light on contemporary evidence on the prognostic impact of CMD. Finally, we will provide an overview of novel emerging therapeutic strategies for CMD.
Collapse
Affiliation(s)
| | - Giorgio Benatti
- Division of Cardiology, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Andrea Denegri
- Division of Cardiology, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Davide Donelli
- Division of Cardiology, University of Parma, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Marco Covani
- Division of Cardiology, University of Parma, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Mattia De Gregorio
- Division of Cardiology, University of Parma, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Gabriella Dallaglio
- Division of Cardiology, University of Parma, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Rebecca Navacchi
- Division of Cardiology, University of Parma, Parma University Hospital, 14 - 43126 Parma, Italy
| | - Giampaolo Niccoli
- Division of Cardiology, University of Parma, Parma University Hospital, 14 - 43126 Parma, Italy
- Division of Cardiology, Parma University Hospital, 14 - 43126 Parma, Italy
| |
Collapse
|
27
|
Di X, Li Y, Wei J, Li T, Liao B. Targeting Fibrosis: From Molecular Mechanisms to Advanced Therapies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410416. [PMID: 39665319 PMCID: PMC11744640 DOI: 10.1002/advs.202410416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/27/2024] [Indexed: 12/13/2024]
Abstract
As the final stage of disease-related tissue injury and repair, fibrosis is characterized by excessive accumulation of the extracellular matrix. Unrestricted accumulation of stromal cells and matrix during fibrosis impairs the structure and function of organs, ultimately leading to organ failure. The major etiology of fibrosis is an injury caused by genetic heterogeneity, trauma, virus infection, alcohol, mechanical stimuli, and drug. Persistent abnormal activation of "quiescent" fibroblasts that interact with or do not interact with the immune system via complicated signaling cascades, in which parenchymal cells are also triggered, is identified as the main mechanism involved in the initiation and progression of fibrosis. Although the mechanisms of fibrosis are still largely unknown, multiple therapeutic strategies targeting identified molecular mechanisms have greatly attenuated fibrotic lesions in clinical trials. In this review, the organ-specific molecular mechanisms of fibrosis is systematically summarized, including cardiac fibrosis, hepatic fibrosis, renal fibrosis, and pulmonary fibrosis. Some important signaling pathways associated with fibrosis are also introduced. Finally, the current antifibrotic strategies based on therapeutic targets and clinical trials are discussed. A comprehensive interpretation of the current mechanisms and therapeutic strategies targeting fibrosis will provide the fundamental theoretical basis not only for fibrosis but also for the development of antifibrotic therapies.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
28
|
Yao J, Zhang Y, Wang Z, Chen Y, Shi X. Maintenance of Cardiac Microenvironmental Homeostasis: A Joint Battle of Multiple Cells. J Cell Physiol 2025; 240:e31496. [PMID: 39632594 DOI: 10.1002/jcp.31496] [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: 07/02/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Various cells such as cardiomyocytes, fibroblasts and endothelial cells constitute integral components of cardiac tissue. The health and stability of cardiac ecosystem are ensured by the action of a certain type of cell and the intricate interactions between multiple cell types. The dysfunctional cells exert a profound impact on the development of cardiovascular diseases by involving in the pathological process. In this paper, we introduce the dynamic activity, cell surface markers as well as biological function of the various cells in the heart. Besides, we discuss the multiple signaling pathways involved in the cardiac injury including Hippo/YAP, TGF-β/Smads, PI3K/Akt, and MAPK signaling. The complexity of different cell types poses a great challenge to the disease treatment. By characterizing the roles of various cell types in cardiovascular diseases, we sought to discuss the potential strategies for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Youtao Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ziwen Wang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| |
Collapse
|
29
|
Lu W, Teoh A, Waters M, Haug G, Shakeel I, Hassan I, Shahzad AM, Callerfelt AKL, Piccari L, Sohal SS. Pathology of idiopathic pulmonary fibrosis with particular focus on vascular endothelium and epithelial injury and their therapeutic potential. Pharmacol Ther 2025; 265:108757. [PMID: 39586361 DOI: 10.1016/j.pharmthera.2024.108757] [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: 05/27/2024] [Revised: 10/15/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) remains a challenging disease with no drugs available to change the trajectory. It is a condition associated with excessive and highly progressive scarring of the lungs with remodelling and extracellular matrix deposition. It is a highly "destructive" disease of the lungs. The diagnosis of IPF is challenging due to continuous evolution of the disease, which also makes early interventions very difficult. The role of vascular endothelial cells has not been explored in IPF in great detail. We do not know much about their contribution to arterial or vascular remodelling, extracellular matrix changes and contribution to pulmonary hypertension and lung fibrosis in general. Endothelial to mesenchymal transition appears to be central to such changes in IPF. Similarly, for epithelial changes, the process of epithelial to mesenchymal transition seem to be the key both for airway epithelial cells and type-2 pneumocytes. We focus here on endothelial and epithelial cell changes and its contributions to IPF. In this review we revisit the pathology of IPF, mechanistic signalling pathways, clinical definition, update on diagnosis and new advances made in treatment of this disease. We discuss ongoing clinical trials with mode of action. A multidisciplinary collaborative approach is needed to understand this treacherous disease for new therapeutic targets.
Collapse
Affiliation(s)
- Wenying Lu
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Alan Teoh
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Maddison Waters
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania 7250, Australia
| | - Greg Haug
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania 7250, Australia
| | - Ilma Shakeel
- Centre For Interdisciplinary Research In Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Imtaiyaz Hassan
- Centre For Interdisciplinary Research In Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Affan Mahmood Shahzad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; Medical School, Oceania University of Medicine, Apia, Samoa
| | | | - Lucilla Piccari
- Department of Pulmonology, Hospital del Mar, Barcelona, Spain
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia.
| |
Collapse
|
30
|
Li Y. Novel Therapeutic Strategies Targeting Fibroblasts to Improve Heart Disease. J Cell Physiol 2025; 240:e31504. [PMID: 39690827 DOI: 10.1002/jcp.31504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Cardiac fibrosis represents the terminal pathological manifestation of various heart diseases, with the formation of fibroblasts playing a pivotal role in this process. Consequently, targeting the formation and function of fibroblasts holds significant potential for improving outcomes in heart disease. Recent research reveals the considerable potential of fibroblasts in ameliorating cardiac conditions, demonstrating different functional characteristics at various time points and spatial locations. Therefore, precise modulation of fibroblast activity may offer an effective approach for treating cardiac fibrosis and achieving targeted therapeutic outcomes. In this review, we focus on the fate and inhibition of fibroblasts, analyze their dynamic changes in cardiac diseases, and propose a framework for identifying markers of fibroblast activation mechanisms and selecting optimal time windows for therapeutic intervention. By synthesizing research findings in these areas, we aim to provide new strategies and directions for the precise treatment of fibroblasts in cardiac diseases.
Collapse
Affiliation(s)
- Yujuan Li
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
31
|
Ding JY, Meng TT, Du RL, Song XB, Li YX, Gao J, Ji R, He QY. Bibliometrics of trends in global research on the roles of stem cells in myocardial fibrosis therapy. World J Stem Cells 2024; 16:1086-1105. [PMID: 39734477 PMCID: PMC11669986 DOI: 10.4252/wjsc.v16.i12.1086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Myocardial fibrosis, a condition linked to several cardiovascular diseases, is associated with a poor prognosis. Stem cell therapy has emerged as a potential treatment option and the application of stem cell therapy has been studied extensively. However, a comprehensive bibliometric analysis of these studies has yet to be conducted. AIM To map thematic trends, analyze research hotspots, and project future directions of stem cell-based myocardial fibrosis therapy. METHODS We conducted a bibliometric and visual analysis of studies in the Web of Science Core Collection using VOSviewer and Microsoft Excel. The dataset included 1510 articles published between 2001 and 2024. Countries, organizations, authors, references, keywords, and co-citation networks were examined to identify evolving research trends. RESULTS Our findings revealed a steady increase in the number of publications, with a projected increase to over 200 publications annually by 2030. Initial research focused on stem cell-based therapy, particularly for myocardial infarction and heart failure. More recently, there has been a shift toward cell-free therapy, involving extracellular vesicles, exosomes, and microRNAs. Key research topics include angiogenesis, inflammation, apoptosis, autophagy, and oxidative stress. CONCLUSION This analysis highlights the evolution of stem cell therapies for myocardial fibrosis, with emerging interest in cell-free approaches. These results are expected to guide future scientific exploration and decision-making.
Collapse
Affiliation(s)
- Jing-Yi Ding
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Tian-Tian Meng
- Department of Rehabilitation, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100071, China
| | - Ruo-Lin Du
- Department of Emergency Medicine, South Branch of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xin-Bin Song
- Department of Intensive Care Unit, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian 463000, Henan Province, China
| | - Yi-Xiang Li
- Department of Chinese Medicine, The Third People's Hospital of Henan Province, Zhengzhou 450000 Henan Province, China
| | - Jing Gao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ran Ji
- Department of Intensive Care Unit, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing-Yong He
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
32
|
Wei B, Huang J, Zhang Y, Hu X, Ma C, Li Y, Chen P. Restoration of RECK expression attenuates liver fibrosis induced by carbon tetrachloride through the Nrf2-MMP9 axis. Int Immunopharmacol 2024; 143:113475. [PMID: 39476567 DOI: 10.1016/j.intimp.2024.113475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
Liver fibrosis is a reversible process that can be delayed or even reversed through appropriate intervention during its development. The protein RECK, encoded by the Reck gene, regulates matrix metalloproteinase (MMP) activity and plays a crucial role in extracellular matrix (ECM) degradation and remodeling. Reduced RECK expression is found in various fibrotic tissues. However, the impact of restoring RECK expression on the development and progression of liver fibrosis has not yet been determined. This study found that the restoration of RECK expression attenuated TGF-β1-induced hepatic stellate cell (HSC) activation and mitigated carbon tetrachloride (CCl4)-induced acute liver injury. In a mouse model of liver fibrosis induced by CCl4, restoration of RECK expression reduced the degree of fibrosis, collagen deposition, and level of oxidative stress. RECK competes with Nrf2 for binding to Keap1, resulting in a decrease in the degradation of Nrf2 by Keap1 and an increase in the accumulation of Nrf2 in the cytoplasm. Under oxidative stress conditions, Nrf2 can be translocated to the nucleus for expression, initiating an antioxidant stress response, furthermore, Nrf2 can also activate MMP-9 and degrade the over-deposited collagen, thereby achieving the effect of alleviating liver fibrosis. Our study reveals a novel mechanism by which restoration of RECK expression ameliorates liver fibrosis, providing a promising target for combating liver fibrosis.
Collapse
Affiliation(s)
- Bizhen Wei
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Jing Huang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yu Zhang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Xiuxiu Hu
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Cao Ma
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yiping Li
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China.
| | - Pingsheng Chen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
33
|
An N, Yang F, Zhang G, Jiang Y, Liu H, Gao Y, Li Y, Ji P, Shang H, Xing Y. Single-cell RNA sequencing reveals the contribution of smooth muscle cells and endothelial cells to fibrosis in human atrial tissue with atrial fibrillation. Mol Med 2024; 30:247. [PMID: 39701940 DOI: 10.1186/s10020-024-00999-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/13/2024] [Indexed: 12/21/2024] Open
Abstract
AIMS Atrial fibrillation (AF) has high mortality and morbidity rates. However, the intracellular molecular complexity of the atrial tissue of patients with AF has not been adequately assessed. METHODS AND RESULTS We investigated the cellular heterogeneity of human atrial tissue and changes in differentially expressed genes between cells using single-cell RNA sequencing, fluorescence in situ hybridization, intercellular communication, and cell trajectory analysis. Using genome-wide association studies (GWAS) and proteomics, we discovered cell types enriched for AF susceptibility genes. We discovered eight different cell types, which were further subdivided into 23 subpopulations. In AF, the communication strength between smooth muscle cells (SMCs) and fibroblast (FB) 3 cells increased and the relevant signaling pathways were quite similar. Subpopulations of endothelial cells (ECs) are mainly involved in fibrosis through TXNDC5 and POSTN. AF susceptibility genes revealed by GWAS were especially enriched in neuronal and epicardial cells, FB3, and lymphoid (Lys) cells, whereas proteomic sequencing differential proteins were concentrated in FB3 cells and SMCs. CONCLUSIONS This study provides a cellular landscape based on the atrial tissue of patients with AF and highlights intercellular changes and differentially expressed genes that occur during the disease process. A thorough description of the cellular populations involved in AF will facilitate the identification of new cell-based interventional targets with direct functional significance for the treatment of human disease.
Collapse
Affiliation(s)
- Na An
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Fan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
| | - Yuchen Jiang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China
| | - Haoqi Liu
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China
| | - Yingjian Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Peifeng Ji
- Computational Genomics Lab, Beijing Institutes of Life Science, Chinese Academy of Sciences, No. 5, Yard 1, Beichen West Road, Chaoyang District, Beijing, 100101, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, 100700, China.
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, No.5 Beixian'ge Street, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
34
|
Li H, Zhang S, Huang X, Zhang Z, Liu K, Wang QD, Chen AF, Lui KO, Sun K, Zhou B. Genetic recording of transient endothelial activation in distinct alveolar capillary cells during pulmonary fibrosis. Cell Discov 2024; 10:119. [PMID: 39622809 PMCID: PMC11612399 DOI: 10.1038/s41421-024-00745-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 10/22/2024] [Indexed: 12/06/2024] Open
Affiliation(s)
- Hongxin Li
- CAS CEMCS-CUHK Joint Laboratories for Cardiovascular Sciences, New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Shaohua Zhang
- CAS CEMCS-CUHK Joint Laboratories for Cardiovascular Sciences, New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuzhen Huang
- CAS CEMCS-CUHK Joint Laboratories for Cardiovascular Sciences, New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhenqian Zhang
- CAS CEMCS-CUHK Joint Laboratories for Cardiovascular Sciences, New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kuo Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kathy O Lui
- CAS CEMCS-CUHK Joint Laboratories for Cardiovascular Sciences, Department of Chemical Pathology; and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories for Cardiovascular Sciences, New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
35
|
Masciale V, Banchelli F, Grisendi G, Samarelli AV, Raineri G, Rossi T, Zanoni M, Cortesi M, Bandini S, Ulivi P, Martinelli G, Stella F, Dominici M, Aramini B. The molecular features of lung cancer stem cells in dedifferentiation process-driven epigenetic alterations. J Biol Chem 2024; 300:107994. [PMID: 39547513 PMCID: PMC11714729 DOI: 10.1016/j.jbc.2024.107994] [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: 03/23/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Cancer stem cells (CSCs) may be dedifferentiated somatic cells following oncogenic processes, representing a subpopulation of cells able to promote tumor growth with their capacities for proliferation and self-renewal, inducing lineage heterogeneity, which may be a main cause of resistance to therapies. It has been shown that the "less differentiated process" may have an impact on tumor plasticity, particularly when non-CSCs may dedifferentiate and become CSC-like. Bidirectional interconversion between CSCs and non-CSCs has been reported in other solid tumors, where the inflammatory stroma promotes cell reprogramming by enhancing Wnt signaling through nuclear factor kappa B activation in association with intracellular signaling, which may induce cells' pluripotency, the oncogenic transformation can be considered another important aspect in the acquisition of "new" development programs with oncogenic features. During cell reprogramming, mutations represent an initial step toward dedifferentiation, in which tumor cells switch from a partially or terminally differentiated stage to a less differentiated stage that is mainly manifested by re-entry into the cell cycle, acquisition of a stem cell-like phenotype, and expression of stem cell markers. This phenomenon typically shows up as a change in the form, function, and pattern of gene and protein expression, and more specifically, in CSCs. This review would highlight the main epigenetic alterations, major signaling pathways and driver mutations in which CSCs, in tumors and specifically, in lung cancer, could be involved, acting as key elements in the differentiation/dedifferentiation process. This would highlight the main molecular mechanisms which need to be considered for more tailored therapies.
Collapse
Affiliation(s)
- Valentina Masciale
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Federico Banchelli
- Department of Statistical Sciences "Paolo Fortunati", Alma Mater Studiorum- University of Bologna, Bologna, Italy
| | - Giulia Grisendi
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Anna Valeria Samarelli
- Laboratory of and Respiratory Medicine, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Giulia Raineri
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michele Zanoni
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Michela Cortesi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Martinelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Franco Stella
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Massimo Dominici
- Laboratory of Cellular Therapies, Department of Medical and Surgical Sciences for Children & Adults, University Hospital of Modena, Modena, Italy; Division of Oncology, University Hospital of Modena and Reggio Emilia, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Thoracic Surgery Unit, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy.
| |
Collapse
|
36
|
Liu H, Wang C, Wang R, Zhang Y, Jian B, Zhou Z, Wu Z, Liang M. HnRNPA1 Prevents Endothelial-to-mesenchymal Transition-induced VSMC Activation and Neointimal Hyperplasia in Vein Grafts. J Cardiovasc Transl Res 2024; 17:1400-1414. [PMID: 39046653 DOI: 10.1007/s12265-024-10545-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024]
Abstract
Endothelial-to-mesenchymal transition (EndoMT) is associated with neointimal hyperplasia and vein graft failure, and heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) has emerged as a major modulator of EMT. We aimed to investigate the functional consequence of EndoMT in neointimal hyperplasia and the precise role of hnRNPA1 in the regulation of EndoMT and neointimal hyperplasia. We investigated the spatial and temporal distribution characteristics of EndoMT cells in a mouse model of vein graft transplantation. In vitro, we studied the interaction between EndoMT cells and VSMCs, and the underlying mechanism was investigated by cytokine antibody assays. In cultured HUVECs, we studied the effect of hnRNPA1 on EndoMT and the cellular interactions by using siRNA-mediated knockdown and adenovirus-mediated overexpression. We further investigated the role of hnRNPA1 in EndoMT and neointimal hyperplasia in vivo with an AAV-mediated EC-specific hnRNPA1 overexpression murine model. We demonstrated the presence of EndoMT cells during the initial stage of neointimal formation, and that EndoMT cells promoted the proliferation and migration of VSMCs in vitro. Mechanistic studies revealed that EndoMT cells express and secrete a higher level of PDGF-B. Furthermore, we found a regulatory role for hnRNPA1 in EndoMT in vitro and in vivo. Similarly, we found that hnRNPA1 overexpression in ECs reduced the expression and secretion of PDGF-B during EndoMT, effectively inhibiting EndoMT cell-mediated activation of VSMCs in vitro and neointimal formation in vivo. Taken together, these findings indicate that EndoMT cells can activate VSMCs through a paracrine mechanism mediated by hnRNPA1 and lead to neointimal hyperplasia.
Collapse
Affiliation(s)
- Haoliang Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Chaoqun Wang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Rui Wang
- Department of Cardiology, Guangzhou Hospital of Integrated Traditional and West Medicine, Guangzhou, 510080, Guangdong, China
| | - Yi Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Bohao Jian
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Zhuoming Zhou
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| | - Mengya Liang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
37
|
Jiang Z, Sorrentino G, Simsek S, Roelofs JJ, Niessen HW, Krijnen PA. Increased perivascular fibrosis and pro-fibrotic cellular transition in intramyocardial blood vessels in myocardial infarction patients. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 10:100275. [PMID: 39801804 PMCID: PMC11708423 DOI: 10.1016/j.jmccpl.2024.100275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025]
Abstract
Background and objectives Structural and functional changes in the intramyocardial microcirculation increase the risk of myocardial infarction (MI). This study investigated intramyocardial perivascular fibrosis and pro-fibrotic cellular transitions in deceased acute and subacute MI patients to explore their involvement in the pathogenesis of MI. Methods Left ventricular tissue (LV) was obtained from the infarction area of autopsied patients with acute-phase MI (3-6 h; n = 24), subacute-phase MI (5-14 days; n = 12), and noninfarcted controls (n = 14). Perivascular fibrosis and fibroblast activation protein (FAP) expression were quantified using (immuno)histochemistry. Fibroblast-like transitioning of vascular smooth muscle cells (VSMC) and endothelial cells (EC) was quantified using immunofluorescent microscopy. Results Perivascular fibrosis was elevated in acute-phase (77.69 %) and subacute-phase (72.19 %: border zone 95.18 %: infarct core) MI patients (p < 0.0001) compared to controls (61.03 %). FAP expression was higher in both acute-phase (1.46 %) and subacute-phase (18.01 %: border zone 5.67 %: infarct core) compared to controls (0.46 %) (p < 0.05). VSMC fibroblast-like cellular transition (SMA + S100A4+ vessels fraction) was higher in acute-phase (31.96 %) and subacute-phase (21.90 %: border zone; 37.25 %: infarct core) MI compared to controls (8.95 %) (p < 0.05). Similarly, EC fibroblast-like cellular transition (CD31 + S100A4+ area fraction) was increased in acute-phase MI (10.14 %) and subacute-phase MI (8.31 %: border zone 10.15 %: infarct core) compared to controls (2.67 %) (p < 0.05). Conclusion Increased perivascular fibrosis, fibroblast activation and vascular cellular transition in intramyocardial blood vessels of MI patients may contribute to MI development. Further increases of FAP expression and perivascular fibrosis, particularly in subacute-phase infarct cores, suggest MI itself exacerbates fibroblast activation and perivascular fibrosis, theoretically increasing reinfarction risk.
Collapse
Affiliation(s)
- Zhu Jiang
- Department of Pathology, Amsterdam University Medical Center (AUMC), location AMC and VUmc, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
| | - Giulia Sorrentino
- Department of Pathology, Amsterdam University Medical Center (AUMC), location AMC and VUmc, Amsterdam, the Netherlands
| | - Suat Simsek
- Department of Internal Medicine, Northwest Clinics, Alkmaar, the Netherlands
- Department of Internal Medicine, AUMC, location VUmc, Amsterdam, the Netherlands
| | - Joris J.T.H. Roelofs
- Department of Pathology, Amsterdam University Medical Center (AUMC), location AMC and VUmc, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
| | - Hans W.M. Niessen
- Department of Pathology, Amsterdam University Medical Center (AUMC), location AMC and VUmc, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
- Department of Cardiac Surgery, AUMC, location AMC and VUmc, Amsterdam, the Netherlands
| | - Paul A.J. Krijnen
- Department of Pathology, Amsterdam University Medical Center (AUMC), location AMC and VUmc, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, the Netherlands
| |
Collapse
|
38
|
Xu Y, Ma X, Ni W, Zheng L, Lin Z, Lai Y, Yang N, Dai Z, Yao T, Chen Z, Shen L, Wang H, Wang L, Wu Y, Gao W. PKM2-Driven Lactate Overproduction Triggers Endothelial-To-Mesenchymal Transition in Ischemic Flap via Mediating TWIST1 Lactylation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406184. [PMID: 39474980 DOI: 10.1002/advs.202406184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/21/2024] [Indexed: 12/19/2024]
Abstract
The accumulation of lactate is a rising risk factor for patients after flap transplantation. Endothelial-to-mesenchymal transition (EndoMT) plays a critical role in skin fibrosis. Nevertheless, whether lactate overproduction directly contributes to flap necrosis and its mechanism remain unknown. The current study reveals that skin flap mice exhibit enhanced PKM2 and fibrotic response. Endothelial-specific deletion of PKM2 attenuates flap necrosis and ameliorates flap fibrosis in mice. Administration of lactate or overexpressing PKM2 promotes dysfunction of endothelial cells and stimulates mesenchymal-like phenotype following hypoxia. Mechanistically, glycolytic-lactate induces a correlation between Twist1 and p300/CBP, leading to lactylation of Twist1 lysine 150 (K150la). The increase in K150la promotes Twist1 phosphorylation and nuclear translocation and further regulates the transcription of TGFB1, hence inducing fibrosis phenotype. Genetically deletion of endothelial-specific PKM2 in mice diminishes lactate accumulation and Twist1 lactylation, then attenuates EndoMT-associated fibrosis following flap ischemia. The serum lactate levels of flap transplantation patients are elevated and exhibit predictive value for prognosis. This findings suggested a novel role of PKM2-derived lactate in mediating Twist1 lactylation and exacerbates flap fibrosis and ischemia. Inhibition of glycolytic-lactate and Twist1 lactylation reduces flap necrosis and fibrotic response might become a potential therapeutic strategy for flap ischemia.
Collapse
Affiliation(s)
- Yining Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xianhui Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Weiyu Ni
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310006, China
| | - Lin Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhongnan Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yingying Lai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhanqiu Dai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Teng Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310006, China
| | - Zeyang Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Lifeng Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310006, China
| | - Haitao Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310006, China
| | - Long Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yizheng Wu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, 310006, China
| | - Weiyang Gao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, 325027, China
- The Second Clinical Medical College of Wenzhou Medical University, Wenzhou, 325027, China
| |
Collapse
|
39
|
Zhao W, Li K, Tang L, Zhang J, Guo H, Zhou X, Luo M, Liu H, Cui R, Zeng M. Coronary Microvascular Dysfunction and Diffuse Myocardial Fibrosis in Patients With Type 2 Diabetes Using Quantitative Perfusion MRI. J Magn Reson Imaging 2024; 60:2395-2406. [PMID: 38376091 DOI: 10.1002/jmri.29296] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/30/2024] [Accepted: 01/30/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Imaging techniques that quantitatively and automatically measure changes in the myocardial microcirculation in patients with diabetes are lacking. PURPOSE To detect diabetic myocardial microvascular complications using a novel automatic quantitative perfusion MRI technique, and to explore the relationship between myocardial microcirculation dysfunction and fibrosis. STUDY TYPE Prospective. SUBJECTS 101 patients with type 2 diabetes mellitus (T2DM) (53 without and 48 with complications), 20 healthy volunteers. FIELD STRENGTH/SEQUENCE 3.0T; modified Look-Locker inversion-recovery sequence; saturation recovery sequence and dual-bolus technique; segmented fast low-angle shot sequence. ASSESSMENT All participants underwent MRI to determine the rest myocardial blood flow (MBF), stress MBF, myocardial perfusion reserve (MPR), and extracellular volume (ECV), which represents the extent of myocardial fibrosis. STATISTICAL TESTS Kolmogorov-Smirnov test, Shapiro-Wilk test, Kruskal-Wallis H test, Mann-Whitney U test, chi-square test, Spearman correlation coefficient, multivariable linear regression analysis. P < 0.05 was considered statistically significant. RESULTS The rest MBF was not significantly different between the T2DM without complications group (1.1, IQR: 0.9-1.3) and the control group (1.1, 1.0-1.3) (P = 1.000), but it was significantly lower in the T2DM with complications group (0.8, 0.6-1.0) than in both other groups. The stress MBF and MPR were significantly lower in the T2DM without complications group (1.9, 1.5-2.3, and 1.7, 1.4-2.1, respectively) than in the control group (3.0, 2.6-3.5, and 2.7, 2.4-3.1, respectively), and were also significantly lower in the T2DM with complications group (1.1, 0.9-1.4, and 1.4, 1.2-1.8, respectively) than in the T2DM without complications group. A decrease in MBF and MPR were significantly associated with an increase in the ECV. DATA CONCLUSION Quantitative perfusion MRI can evaluate myocardial microcirculation dysfunction. In T2DM, there was a significant decrease in both MBF and MPR compared to healthy controls, with the decrease being significantly different between T2DM with and without complications groups. The decrease of MBF was significantly associated with the development of myocardial fibrosis, as determined by ECV. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Wenjin Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Kang Li
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Leting Tang
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiamin Zhang
- Department of Radiology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Hu Guo
- MR Application, Siemens Healthineers Ltd., Changsha, China
| | - Xiaoyue Zhou
- MR Collaboration, Siemens Healthineers Ltd., Shanghai, China
| | - Meichen Luo
- Circle Cardiovascular Imaging Inc., Calgary, Alberta, Canada
| | - Hongduan Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rongrong Cui
- National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Mu Zeng
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Medical Imaging in Hunan Province, Changsha, China
| |
Collapse
|
40
|
Liu J, Zhou F, Tang Y, Li L, Li L. Progress in Lactate Metabolism and Its Regulation via Small Molecule Drugs. Molecules 2024; 29:5656. [PMID: 39683818 DOI: 10.3390/molecules29235656] [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: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Lactate, once viewed as a byproduct of glycolysis and a metabolic "waste", is now recognized as an energy-providing substrate and a signaling molecule that modulates cellular functions under pathological conditions. The discovery of histone lactylation in 2019 marked a paradigm shift, with subsequent studies revealing that lactate can undergo lactylation with both histone and non-histone proteins, implicating it in the pathogenesis of various diseases, including cancer, liver fibrosis, sepsis, ischemic stroke, and acute kidney injury. Aberrant lactate metabolism is associated with disease onset, and its levels can predict disease outcomes. Targeting lactate production, transport, and lactylation may offer therapeutic potential for multiple diseases, yet a systematic summary of the small molecules modulating lactate and its metabolism in various diseases is lacking. This review outlines the sources and clearance of lactate, as well as its roles in cancer, liver fibrosis, sepsis, ischemic stroke, myocardial infarction, and acute kidney injury, and summarizes the effects of small molecules on lactate regulation. It aims to provide a reference and direction for future research.
Collapse
Affiliation(s)
- Jin Liu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Feng Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yang Tang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Linghui Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ling Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
41
|
Lee SE, Yu SH, Kim IH, Kang YC, Kim Y, Yeo JS, Lim JH, Kwon I, Kim JH, Park SW, Chang MY, Han K, Kim SH, Kim CH. Mitochondrial Transplantation Ameliorates Pulmonary Fibrosis by Suppressing Myofibroblast Activation. Int J Mol Sci 2024; 25:12783. [PMID: 39684495 DOI: 10.3390/ijms252312783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a pulmonary disease characterized by excessive extracellular matrix protein deposition in the lung interstitium, subsequently causing respiratory failure. IPF still has a high medical unmet requirement due to the lack of effective treatments to inhibit disease progression. The etiology of IPF remains unclear, but mitochondrial dysfunction is considered to be associated with IPF development. Therefore, targeting mitochondrial abnormalities would be a promising strategy for treating IPF. Recently, exogenous mitochondrial transplantation has been beneficial for treating mitochondrial dysfunction. The current study aimed to examine the therapeutic effect of mitochondrial transplantation on IPF in vitro and in vivo. Mitochondria were isolated from human umbilical cord mesenchymal stem cells, referred to as PN-101. Human lung fibroblasts and human bronchial epithelial cells were exposed to transforming growth factor-β, followed by PN-101 treatment to determine the in vitro efficacy of mitochondrial transplantation. An IPF mouse model established by a single intratracheal instillation of bleomycin was utilized to determine the in vivo efficacy of the intravenously treated mitochondria. PN-101 attenuated mitochondrial damage, inhibited EMC production, and suppressed epithelial-to-mesenchymal transition in vitro. Additionally, intravenous PN-101 administration alleviated bleomycin-induced fibrotic processes in the IPF mouse model with a therapeutic context. Our data indicate that PN-101 is a novel and potential therapeutic agent for IPF.
Collapse
Affiliation(s)
- Seo-Eun Lee
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Shin-Hye Yu
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - In-Hyeon Kim
- Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Young Cheol Kang
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Yujin Kim
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Jeong Seon Yeo
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Jun Hyeok Lim
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Iksun Kwon
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Je-Hein Kim
- Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Se-Woong Park
- Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
- College of Veterinary Medicine, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Mi-Yoon Chang
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Republic of Korea
- Department of Premedicine, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Kyuboem Han
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| | - Sung-Hwan Kim
- Division of Jeonbuk Advanced Bio Research, Korea Institute of Toxicology, Jeongeup 56212, Republic of Korea
| | - Chun-Hyung Kim
- Paean Biotechnology, Inc., 5 Samil-daero 8-gil, Jung-gu, Seoul 04552, Republic of Korea
| |
Collapse
|
42
|
Zhang J, Wang S, Sun Q, Zhang J, Shi X, Yao M, Chen J, Huang Q, Zhang G, Huang Q, Ai K, Bai Y. Peroxynitrite-Free Nitric Oxide-Embedded Nanoparticles Maintain Nitric Oxide Homeostasis for Effective Revascularization of Myocardial Infarcts. ACS NANO 2024; 18:32650-32671. [PMID: 39545833 DOI: 10.1021/acsnano.4c10118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Revascularization is crucial for treating myocardial infarction (MI). Nitric oxide (NO), at an appropriate concentration, is recognized as an ideal and potent pro-angiogenic factor. However, the application of NO in the treatment of MI is limited. Improper NO supplementation is harmful to revascularization because NO is converted into harmful peroxynitrite (ONOO-) in MI tissues with high reactive oxygen species (ROS) levels. We overcome these obstacles by embedding biliverdin and NO into Prussian blue (PB) nanolattices to obtain an ONOO--free NO-embedded nanomedicine (OFEN). Unlike previous NO donors, OFEN provides NO stably and spontaneously for a longer time (>7 days), which makes it possible to maintain a stable concentration of NO, suitable for angiogenesis, through dose optimization. More importantly, based on the synergy between PB and biliverdin, OFEN converts ROS into beneficial O2 and inhibits the production of ONOO- from the source. OFEN specifically targets MI tissues and achieves sustained and stable NO delivery at the MI site. OFEN effectively promotes revascularization in the MI tissue, significantly reduces myocardial death and fibrosis, and ultimately promotes the complete recovery of cardiac function. Our strategy provides a promising approach for the treatment of myocardial and other ischemic diseases.
Collapse
Affiliation(s)
- Jiaxiong Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Quan Sun
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jian Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Meilian Yao
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jing Chen
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Guogang Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, PR China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Yongping Bai
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
43
|
Zou Y, Shi H, Li Y, Li T, Liu N, Liu B. Heat shock protein 27 downregulation attenuates isoprenaline-induced myocardial fibrosis and diastolic dysfunction by modulating the endothelial-mesenchymal transition. Biochem Pharmacol 2024; 230:116612. [PMID: 39515591 DOI: 10.1016/j.bcp.2024.116612] [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: 05/20/2024] [Revised: 10/11/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Heart failure (HF), an end-stage clinical syndrome secondary to cardiac impairment, significantly affects patients' quality of life and long-term prognosis. Myocardial fibrosis leads to systolic and diastolic dysfunction, and promotes the progression of HF. Several studies involving the modulation of myocardial fibrosis have been conducted in an effort to improve cardiac function. Heat shock protein 27 (HSP27) is a small chaperone protein that is overexpressed in cellular stress states. HSP27 modulates epithelial-mesenchymal transition, playing a crucial role in the pathology of several fibrotic diseases. However, its association with myocardial fibrosis regulation is unknown. This study aimed to investigate the mechanisms by which HSP27 regulates myocardial fibrosis. We created cardiac-specific HSP25 (the murine ortholog of human HSP27) knockout mice and found that HSP25 knockdown inhibited endothelial-mesenchymal transition (EndMT), attenuated myocardial fibrosis, and ameliorated diastolic dysfunction in isoproterenol-induced HF mice via echocardiography, histology, and western bloting. In vitro, HSP27 knockdown attenuated transforming growth factor beta-induced EndMT, whereas HSP27 overexpression promoted EndMT. Furthermore, the SMAD3/SNAIL1 pathway was found to be crucial for HSP27-mediated EndMT regulation. As an essential molecule in EndMT regulation and myocardial fibrosis modulation, HSP27 may hold promise as a therapeutic target for patients with HF.
Collapse
Affiliation(s)
- Yifei Zou
- Department of Cardiology, Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun 130041, China
| | - Henghe Shi
- Department of Cardiology, Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun 130041, China
| | - Yinghao Li
- Department of Cardiology, Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun 130041, China
| | - Tianyi Li
- Department of Cardiology, Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun 130041, China
| | - Ning Liu
- Department of Cardiology, Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun 130041, China.
| | - Bin Liu
- Department of Cardiology, Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun 130041, China.
| |
Collapse
|
44
|
Gui Z, Liu X, Xu Z, Feng D, Hang Z, Zheng M, Chen H, Fei S, Sun L, Tao J, Han Z, Ju X, Gu M, Tan R, Wang Z. Src inhibition modulates AMBRA1-mediated mitophagy to counteract endothelial-to-mesenchymal transition in renal allograft fibrosis. Cell Prolif 2024; 57:e13699. [PMID: 38943534 PMCID: PMC11533082 DOI: 10.1111/cpr.13699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/06/2024] [Accepted: 06/09/2024] [Indexed: 07/01/2024] Open
Abstract
Chronic allograft dysfunction (CAD) poses a significant challenge in kidney transplantation, with renal vascular endothelial-to-mesenchymal transition (EndMT) playing a vital role. While renal vascular EndMT has been verified as an important contributing factor to renal allograft interstitial fibrosis/tubular atrophy in CAD patients, its underlying mechanisms remain obscure. Currently, Src activation is closely linked to organ fibrosis development. Single-cell transcriptomic analysis in clinical patients revealed that Src is a potential pivotal mediator in CAD progression. Our findings revealed a significant upregulation of Src which closely associated with EndMT in CAD patients, allogeneic kidney transplanted rats and endothelial cells lines. In vivo, Src inhibition remarkably alleviate EndMT and renal allograft interstitial fibrosis in allogeneic kidney transplanted rats. It also had a similar antifibrotic effect in two endothelial cell lines. Mechanistically, the knockout of Src resulted in an augmented AMBRA1-mediated mitophagy in endothelial cells. We demonstrate that Src knockdown upregulates AMBRA1 level and activates mitophagy by stabilizing Parkin's ubiquitination levels and mitochondrial translocation. Subsequent experiments demonstrated that the knockdown of the Parkin gene inhibited mitophagy in endothelial cells, leading to increased production of Interleukin-6, thereby inducing EndMT. Consequently, our study underscores Src as a critical mediator of renal vascular EndMT and allograft interstitial fibrosis, exerting its impact through the regulation of AMBRA1/Parkin-mediated mitophagy.
Collapse
Affiliation(s)
- Zeping Gui
- Department of UrologyThe Second Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Xuzhong Liu
- Department of UrologyHuai'an First People's Hospital, Nanjing Medical UniversityHuai'anChina
- Department of UrologyAffiliated Clinical College of Xuzhou Medical UniversityHuai'anChina
| | - Zhen Xu
- Department of UrologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhouChina
| | - Dengyuan Feng
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Zhou Hang
- Department of UrologyThe Second Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Ming Zheng
- Department of UrologyThe Second Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Hao Chen
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Shuang Fei
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Li Sun
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Jun Tao
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Zhijian Han
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Xiaobin Ju
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Min Gu
- Department of UrologyThe Second Affiliated Hospital with Nanjing Medical UniversityNanjingChina
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Ruoyun Tan
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| | - Zijie Wang
- Department of UrologyThe First Affiliated Hospital with Nanjing Medical UniversityNanjingChina
| |
Collapse
|
45
|
Yan W, Wu H, Wu Y, Gao Z, Li Z, Zhao F, Cao C, Wang J, Cheng J, Hu X, Ao Y. Exercise Induced Endothelial Mesenchymal Transition (EndMT) Facilitates Meniscal Fibrocartilage Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403788. [PMID: 39344749 PMCID: PMC11600215 DOI: 10.1002/advs.202403788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/01/2024] [Indexed: 10/01/2024]
Abstract
The meniscus is a semilunar wedge-shaped fibrocartilage tissue within the knee joint that is important for withstanding mechanical shock during joint motion. The intrinsic healing capacity of meniscus tissue is very limited, which makes meniscectomy the primary treatment method in the clinic. An effective translational strategy for regenerating the meniscus after total or subtotal meniscectomy, particularly for extensive meniscal lesions or degeneration, is yet to be developed. The present study demonstrates that the endothelial mesenchymal transition (EndMT) contributes to meniscal regeneration. The mechanical stimulus facilitated EndMT by activating TGF-β2 signaling. A handheld bioprinter system to intraoperatively fabricate a porous meniscus scaffold according to the resected meniscus tissue is developed; this can simplify the scaffold fabrication procedure and period. The transplantation of a porous meniscus scaffold combined with a postoperative regular exercise stimulus facilitated the regeneration of anisotropic meniscal fibrocartilaginous tissue and protected the joint cartilage from degeneration in an ovine subtotal meniscectomy model. Single-cell RNA sequencing and immunofluorescence co-staining analyses further confirmed the occurrence of EndMT during meniscal regeneration. EndMT-transformed cells gave rise to fibrochondrocytes, subsequently contributing to meniscal fibrocartilage regeneration. Thus, an efficient translational strategy to facilitate meniscal regeneration is developed.
Collapse
Affiliation(s)
- Wenqiang Yan
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Haoda Wu
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Yue Wu
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Zeyuan Gao
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Zong Li
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Fengyuan Zhao
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Chenxi Cao
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Jianquan Wang
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Jin Cheng
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Xiaoqing Hu
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| | - Yingfang Ao
- Department of Sports MedicinePeking University Third HospitalInstitute of Sports Medicine of Peking UniversityBeijing100191China
- Beijing Key Laboratory of Sports InjuriesBeijing100191China
- Engineering Research Center of Sports Trauma Treatment Technology and DevicesMinistry of EducationBeijing100191China
| |
Collapse
|
46
|
Huang L. The role of IL-17 family cytokines in cardiac fibrosis. Front Cardiovasc Med 2024; 11:1470362. [PMID: 39502194 PMCID: PMC11534612 DOI: 10.3389/fcvm.2024.1470362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/29/2024] [Indexed: 11/08/2024] Open
Abstract
Myocardial fibrosis is a common pathological feature in various cardiovascular diseases including myocardial infarction, heart failure, and myocarditis. Generally, persistent myocardial fibrosis correlates with poor prognosis and ranks among the leading causes of death globally. Currently, there is no effective treatment for myocardial fibrosis, partly due to its unclear pathogenic mechanism. Increasing studies have shown IL-17 family cytokines are strongly associated with the initiation and propagation of myocardial fibrosis. This review summarizes the expression, action, and signal transduction mechanisms of IL-17, focusing on its role in fibrosis associated with cardiovascular diseases such as myocardial infarction, heart failure, hypertension, diabetes, and myocarditis. It also discusses its potential as a therapeutic target, offering new insights for the clinical treatment of myocardial fibrosis.
Collapse
Affiliation(s)
- Liqing Huang
- Three Gorges University Hospital of Traditional Chinese Medicine & Yichang Hospital of Traditional Chinese Medicine, Yichang, China
| |
Collapse
|
47
|
Hashiesh HM, Azimullah S, Nagoor Meeran MF, Saraswathiamma D, Arunachalam S, Jha NK, Sadek B, Adeghate E, Sethi G, Albawardi A, Al Marzooqi S, Ojha S. Cannabinoid 2 Receptor Activation Protects against Diabetic Cardiomyopathy through Inhibition of AGE/RAGE-Induced Oxidative Stress, Fibrosis, and Inflammasome Activation. J Pharmacol Exp Ther 2024; 391:241-257. [PMID: 38955492 DOI: 10.1124/jpet.123.002037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
Oxidative stress, fibrosis, and inflammasome activation from advanced glycation end product (AGE)-receptor of advanced glycation end product (RAGE) interaction contribute to diabetic cardiomyopathy (DCM) formation and progression. Our study revealed the impact of β-caryophyllene (BCP) on activating cannabinoid type 2 receptors (CB2Rs) against diabetic complication, mainly cardiomyopathy and investigated the underlying cell signaling pathways in mice. The murine model of DCM was developed by feeding a high-fat diet with streptozotocin injections. After the development of diabetes, the animals received a 12-week oral BCP treatment at a dose of 50 mg/kg/body weight. BCP treatment showed significant improvement in glucose tolerance and insulin resistance and enhanced serum insulin levels in diabetic animals. BCP treatment effectively reversed the heart remodeling and restored the phosphorylated troponin I and sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a expression. Ultrastructural examination showed reduced myocardial cell injury in DCM mice treated with BCP. The preserved myocytes were found to be associated with reduced expression of AGE/RAGE in DCM mice hearts. BCP treatment mitigated oxidative stress by inhibiting expression of NADPH oxidase 4 and activating phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling. Also, BCP suppressed cardiac fibrosis and endothelial-to-mesenchymal transition in DCM mice by inhibiting transforming growth factor β (TGF-β)/suppressor of mothers against decapentaplegic (Smad) signaling. Further, BCP treatment suppressed nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome activation in DCM mice and alleviated cellular injury to the pancreatic tissues evidenced by significant elevation of the number of insulin-positive cells. To demonstrate a CB2R-dependent mechanism of BCP, another group of DCM mice were pretreated with AM630, a CB2R antagonist. AM630 was observed to abrogate the beneficial effects of BCP in DCM mice. Taken together, BCP demonstrated the potential to protect the myocardium and pancreas of DCM mice mediating CB2R-dependent mechanisms. SIGNIFICANCE STATEMENT: BCP, a CB2R agonist, shows protection against DCM. BCP attenuates oxidative stress, inflammation, and fibrosis in DCM via activating CB2Rs. BCP mediating CB2R activation favorably modulates AGE/RAGE, PI3K/AKT/Nrf2β and TGF-β/Smad and (NLRP3) inflammasome in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Hebaallah Mamdouh Hashiesh
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Sheikh Azimullah
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Mohamed Fizur Nagoor Meeran
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Dhanya Saraswathiamma
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Seenipandi Arunachalam
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Niraj Kumar Jha
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Bassem Sadek
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Ernest Adeghate
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Gautam Sethi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Alia Albawardi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Saeeda Al Marzooqi
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| | - Shreesh Ojha
- Departments of Pharmacology and Therapeutics (H.M.H., Sh.A., M.F.N.M., Se.A., B.S., S.O.), Pathology (D.S., A.A., S.A.M.), and Anatomy (E.A.), College of Medicine and Health Sciences, and Zayed Bin Sultan Center for Health Sciences (S.O.), United Arab Emirates University, Al Ain, United Arab Emirates; Department of Pharmacology and Toxicology, Helwan University, Cairo, Egypt (H.M.H.); Department of Pharmaceutical Biosciences, Research; Drug Safety and Toxicology, Uppsala Biomedicines Centrum BMC, UPPSALA, Sweden (Sh.A.); Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, India (N.K.J.); School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, India (N.K.J.); and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (G.S.)
| |
Collapse
|
48
|
Wang J, Zheng C, Lu J, Xu X, Xiang G, Li J, Zhang J, Mu X, Lu Q. The mechanism of MMP14-positive tumor-associated fibroblast subsets in inhibiting PD-1 immunotherapy for esophageal cancer through exosomal tsRNA-10522. Funct Integr Genomics 2024; 24:186. [PMID: 39377944 PMCID: PMC11461773 DOI: 10.1007/s10142-024-01447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Esophageal cancer (EC) continues to pose a significant health risk. Cancer-associated fibroblasts (CAFs), an essential part of the tumor microenvironment (TME), are viewed as potential therapeutic targets. However, their role in tumor mechanisms specific to esophageal cancer remains to be elucidated. This study identified MMP14+ CAFs and MMP14- CAFs using immunofluorescence staining. The cytotoxic activity of CD8 T cells was assessed via western blot and ELISA. Using a transwell test, the migratory potential of MMP14+ CAFs was evaluated. Using flow cytometry, apoptosis was found in the esophageal squamous cell carcinoma cell line KYSE30. To determine the important tsRNAs released by MMP14+ CAFs, tsRNA-seq was used. Two subgroups of EC receiving PD-1 immunotherapy were identified by our research: MMP14+ CAFs and MMP14- CAFs. MMP14+ CAFs showed improved migratory capacity and released more inflammatory factors linked to cancer. Through exosomes, these CAFs may prevent anti-PD-1-treated CD8 T cells from being cytotoxic. Furthermore, exosomal tsRNA from MMP14+ CAFs primarily targeted signaling pathways connected with cancer. Notably, it was discovered that tsRNA-10522 plays a critical role within inhibiting CD8 T cell tumor cell death. The tumor cell killing of CD8 T cells by exosomal tsRNA-10522 is inhibited by a subgroup of cells called MMP14+ CAFs inside the EC microenvironment during PD-1 immunotherapy. This reduces the effectiveness of PD-1 immunotherapy for EC. Our findings demonstrate the inhibitory function of MMP14+ CAFs within EC receiving PD-1 immunotherapy, raising the prospect that MMP14+ CAFs might serve as predictive indicators in EC receiving PD-1 immunotherapy.
Collapse
Affiliation(s)
- Juzheng Wang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
- Department of Thoracic Surgery, The First People's Hospital of Xianyang, Xianyang, 712000, Shaanxi, China
| | - Chunlong Zheng
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, NO. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Jiayu Lu
- Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Xinyao Xu
- College of Life Sciences, Northwest University, 229 Taibai North Road, Beilin District, Xi'an, 710069, Shaanxi, China
| | - Guangyu Xiang
- Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiahe Li
- Basic Medical College, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Jipeng Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, NO. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Xiaorong Mu
- Department of Pathology, Department of Pharmacy, Tangdu Hospital, Air Force Medical University, Xi'an, 710039, Shaanxi, China.
| | - Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, NO. 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
49
|
Wang Z, Li L, Yang S, Li Z, Zhang P, Shi R, Zhou X, Tang X, Li Q. Possible mechanisms of SARS-CoV-2-associated myocardial fibrosis: reflections in the post-pandemic era. Front Microbiol 2024; 15:1470953. [PMID: 39444690 PMCID: PMC11497467 DOI: 10.3389/fmicb.2024.1470953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Since December 2019, coronavirus disease 2019 (COVID-19) has been spreading worldwide with devastating immediate or long-term effects on people's health. Although the lungs are the primary organ affected by COVID-19, individuals infected with SARS-CoV-2 also develop systemic lesions involving multiple organs throughout the body, such as the cardiovascular system. Emerging evidence reveals that COVID-19 could generate myocardial fibrosis, termed "COVID-19-associated myocardial fibrosis." It can result from the activation of fibroblasts via the renin-angiotensin-aldosterone system (RAAS), transforming growth factor-β1 (TGF-β1), microRNAs, and other pathways, and can also occur in other cellular interactions with SARS-CoV-2, such as immunocytes, endothelial cells. Nonetheless, to gain a more profound insight into the natural progression of COVID-19-related myocardial fibrosis, additional investigations are necessary. This review delves into the underlying mechanisms contributing to COVID-19-associated myocardial fibrosis while also examining the antifibrotic potential of current COVID-19 treatments, thereby offering guidance for future clinical trials of these medications. Ultimately, we propose future research directions for COVID-19-associated myocardial fibrosis in the post-COVID-19 era, such as artificial intelligence (AI) telemedicine. We also recommend that relevant tests be added to the follow-up of COVID-19 patients to detect myocardial fibrosis promptly.
Collapse
Affiliation(s)
- Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Luwei Li
- Department of Pediatric Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Third Clinical Medical College of Zhengzhou University, Zhengzhou, China
| | - Shuai Yang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengpeng Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xing Zhou
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojuan Tang
- Department of Plastic and Reconstructive Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Li
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
50
|
Stock AT, Parsons S, Hansen JA, D'Silva DB, Starkey G, Fayed A, Lim XY, D'Costa R, Gordon CL, Wicks IP. mTOR signalling controls the formation of smooth muscle cell-derived luminal myofibroblasts during vasculitis. EMBO Rep 2024; 25:4570-4593. [PMID: 39271773 PMCID: PMC11467406 DOI: 10.1038/s44319-024-00251-1] [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: 12/19/2023] [Revised: 08/02/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The accumulation of myofibroblasts within the intimal layer of inflamed blood vessels is a potentially catastrophic complication of vasculitis, which can lead to arterial stenosis and ischaemia. In this study, we have investigated how these luminal myofibroblasts develop during Kawasaki disease (KD), a paediatric vasculitis typically involving the coronary arteries. By performing lineage tracing studies in a murine model of KD, we reveal that luminal myofibroblasts develop independently of adventitial fibroblasts and endothelial cells, and instead derive from smooth muscle cells (SMCs). Notably, the emergence of SMC-derived luminal myofibroblasts-in both mice and patients with KD, Takayasu's arteritis and Giant Cell arteritis-coincided with activation of the mechanistic target of rapamycin (mTOR) signalling pathway. Moreover, SMC-specific deletion of mTOR signalling, or pharmacological inhibition, abrogated the emergence of luminal myofibroblasts. Thus, mTOR is an intrinsic and essential regulator of luminal myofibroblast formation that is activated in vasculitis patients and therapeutically tractable. These findings provide molecular insight into the pathogenesis of coronary artery stenosis and identify mTOR as a therapeutic target in vasculitis.
Collapse
Affiliation(s)
| | - Sarah Parsons
- Department of Forensic Medicine, Monash University, Melbourne, VIC, 3006, Australia
- Victorian Institute of Forensic Medicine, Melbourne, VIC, 3006, Australia
| | | | | | - Graham Starkey
- Liver & Intestinal Transplant Unit, Austin Health, Melbourne, VIC, 3084, Australia
- Department of Surgery, The University of Melbourne, Austin Health, Melbourne, VIC, 3084, Australia
| | - Aly Fayed
- Department of Surgery, Austin Health, Melbourne, VIC, 3084, Australia
| | - Xin Yi Lim
- Department of Infectious Diseases, Austin Health, Melbourne, VIC, 3084, Australia
| | - Rohit D'Costa
- DonateLife Victoria, Carlton, VIC, 3053, Australia
- Department of Intensive Care Medicine, Melbourne Health, Melbourne, VIC, 3084, Australia
| | - Claire L Gordon
- Department of Infectious Diseases, Austin Health, Melbourne, VIC, 3084, Australia
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, 3052, Australia
- North Eastern Public Health Unit, Austin Health, Melbourne, VIC, 3084, Australia
| | - Ian P Wicks
- WEHI, Melbourne, VIC, 3052, Australia.
- Rheumatology Unit, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
- University of Melbourne, Department of Medical Biology, Melbourne, VIC, 3052, Australia.
| |
Collapse
|