1
|
Zhang M, Kong X, Wu C, Li J, Yang H, Huang L. The role of lactate and lactylation in ischemic cardiomyopathy: Mechanisms and gene expression. Exp Mol Pathol 2025; 141:104957. [PMID: 40020527 DOI: 10.1016/j.yexmp.2025.104957] [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: 05/30/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Ischemic cardiomyopathy (ICM) is a significant global public health issue, with its pathophysiology encompassing atherosclerotic plaque formation, thrombosis, hypoperfusion, ischemic cell death, and left ventricular remodeling. Lactate is not only regarded as an energy metabolite but also acts as a signaling molecule that influences various physiological processes, regulating metabolism and muscle contraction. Lactylation, an emerging epigenetic modification, affects protein functionality and gene expression through the P300 enzyme. In ICM, lactate accumulation leads to pH imbalance and myocardial cell dysfunction, impacting cellular signaling. This paper will analyze the role of lactylation in ICM, focusing on coronary artery disease (ASCVD) and myocardial infarction (MI). It will also explore the differential expression and immunological characteristics of lactylation-related genes in normal and ICM tissues, providing potential targets for future research.
Collapse
Affiliation(s)
- Mei Zhang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Xue Kong
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China
| | - Chenlu Wu
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiuhong Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hui Yang
- Department of Cardiology, the Second Xiangya Hospital of Central South University, Changsha, China.
| | - Lingzhi Huang
- Department of Clinical Nursing, The Second Xiangya Hospital of Central South University, Changsha, China; Xiangya School of Nursing, Central South University, Changsha, China.
| |
Collapse
|
2
|
Yilmaz E, Yilmaz D, Yildiz CG, Cacan E. Upregulation of the MAP2K4 gene triggers endothelial-mesenchymal transition in COVID-19. Mol Biol Rep 2025; 52:180. [PMID: 39888478 DOI: 10.1007/s11033-025-10289-6] [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: 09/22/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND SARS-CoV-2 infection is marked by an excessive inflammatory response, leading to elevated production of pro-inflammatory cytokines through activation of intracellular pathways like mitogen-activated protein kinase (MAPK). Viruses can use the MAPK signaling pathway to their advantage, but the relationship of this pathway to the severe SARS-CoV-2 period has not been fully elucidated. MAP2K4 is involved in the MAPK signaling pathway and affects cellular processes such as cell-cell junction, cell proliferation, differentiation and apoptosis. METHODS AND RESULTS In this study, we sought to determine the associated biomarkers that are involved in the MAP2K4 pathway and elucidate its possible roles in terms of some clinical features associated with COVID-19. We evaluated the expressions of MAP2K4, SNAI1, SLUG, ZEB1 and E-Cadherin. For this purpose, we prospectively recruited 66 individuals, 39 of whom were women and had a mean age of 65 years. The results revealed that MAP2K4 upregulation increased SNAI1 gene expression level whereas E- Cadherin level was decreased in SARS-CoV-2 positive participants. In addition, negative correlations were determined with PLT, Lymphocyte and CKMB and E- Cadherin levels in positive participants. We also observed a negative correlation between the MAP2K4 and AST, and a positive correlation between SLUG and BUN, ZEB1 and CK. CONCLUSIONS We conclude that SARS-CoV-2 infection triggers fibrosis by increasing MAP2K4 regulation. Additionally, this is the first study to demonstrate the possible contribution of MAP2K4 in influencing COVID-19 clinical features, which may be relevant for identifying COVID-19 positive participants with severe complications.
Collapse
Affiliation(s)
- Esra Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye
| | - Dilek Yilmaz
- Department of Infectious Diseases and Clinical Microbiology, Yozgat City Hospital, Tokat, 66100, Türkiye
| | - Can Gokay Yildiz
- Department of Emergency Medicine, Tokat City Hospital, Tokat, 60200, Türkiye
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Faculty of Art and Science, Tokat Gaziosmanpasa University, Tokat, 60200, Türkiye.
| |
Collapse
|
3
|
Abe S, Inagaki Y, Kokudo T, Miyata A, Nishioka Y, Ichida A, Kaneko J, Akamatsu N, Kawaguchi Y, Hasegawa K. c-Met inhibitor upregulates E-cadherin, which is lost in portal vein tumor thrombus of hepatocellular carcinoma. Hepatol Res 2024; 55:240-249. [PMID: 39367844 DOI: 10.1111/hepr.14120] [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: 03/08/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 10/07/2024]
Abstract
AIM Portal vein tumor thrombus (PVTT) in hepatocellular carcinoma (HCC) is an essential therapeutic and prognostic factor. E-cadherin plays a crucial role in adhesive properties and intercellular interaction in various cancer tissues, including HCC, but the expression profile and functional contribution of E-cadherin in PVTT remain unknown. This study aimed to analyze the expression of E-cadherin in the main tumor tissue and PVTT tissue of HCC, and evaluate the functional roles of E-cadherin in PVTT formation. METHODS A retrospective analysis was performed using the medical records of patients who underwent liver resection for HCC with PVTT, analyzing tissue specimens from 1995 to 2016. E-cadherin expression is evaluated using immunohistochemistry and western blot. The study also uses a c-Met inhibitor to explore its impact on E-cadherin expression in vitro and in vivo using cell lines and a tumor xenograft mouse model. RESULTS The results revealed a reduced E-cadherin expression in PVTT tissue than in the main tumor tissue. The inhibition of c-Met activation, frequently detected in HCC, upregulated E-cadherin expression in HCC cell lines. Furthermore, treatment with c-Met inhibitors induced changes in epithelial morphology, and inhibited migration and invasion of HCC cell lines. CONCLUSIONS This study demonstrates the downregulation of E-cadherin in PVTT, and underscores the potential of c-Met inhibition in upregulating E-cadherin and inhibiting metastatic behavior. Understanding the significance of E-cadherin and c-Met in HCC progression provides a foundation for future clinical investigations into the therapeutic effects of c-Met inhibitors on PVTT in HCC patients.
Collapse
Affiliation(s)
- Satoru Abe
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshinori Inagaki
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Kampo Medicine, Yokohama University of Pharmacy, Yokohama, Kanagawa, Japan
| | - Takashi Kokudo
- Department of Surgery, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akinori Miyata
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yujiro Nishioka
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiko Ichida
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Kaneko
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuhisa Akamatsu
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshikuni Kawaguchi
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kiyoshi Hasegawa
- Hepato-Biliary-Pancreatic Surgery Division and Artificial Organ and Transplantation Division, Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Zhu W, Guo S, Sun J, Zhao Y, Liu C. Lactate and lactylation in cardiovascular diseases: current progress and future perspectives. Metabolism 2024; 158:155957. [PMID: 38908508 DOI: 10.1016/j.metabol.2024.155957] [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: 12/06/2023] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/24/2024]
Abstract
Cardiovascular diseases (CVDs) are often linked to structural and functional impairments, such as heart defects and circulatory dysfunction, leading to compromised peripheral perfusion and heightened morbidity risks. Metabolic remodeling, particularly in the context of cardiac fibrosis and inflammation, is increasingly recognized as a pivotal factor in the pathogenesis of CVDs. Metabolic syndromes further predispose individuals to these conditions, underscoring the need to elucidate the metabolic underpinnings of CVDs. Lactate, a byproduct of glycolysis, is now recognized as a key molecule that connects cellular metabolism with the regulation of cellular activity. The transport of lactate between different cells is essential for metabolic homeostasis and signal transduction. Disruptions to lactate dynamics are implicated in various CVDs. Furthermore, lactylation, a novel post-translational modification, has been identified in cardiac cells, where it influences protein function and gene expression, thereby playing a significant role in CVD pathogenesis. In this review, we summarized recent advancements in understanding the role of lactate and lactylation in CVDs, offering fresh insights that could guide future research directions and therapeutic interventions. The potential of lactate metabolism and lactylation as innovative therapeutic targets for CVD is a promising avenue for exploration.
Collapse
Affiliation(s)
- Wengen Zhu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| | - Siyu Guo
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China
| | - Junyi Sun
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China
| | - Yudan Zhao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430023, PR China.
| | - Chen Liu
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, PR China; Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou 510080, PR China.
| |
Collapse
|
5
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
6
|
Singh B, Cui K, Eisa-Beygi S, Zhu B, Cowan DB, Shi J, Wang DZ, Liu Z, Bischoff J, Chen H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 155:107368. [PMID: 38548093 PMCID: PMC11303600 DOI: 10.1016/j.vph.2024.107368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis, a chronic systemic inflammatory condition, is implicated in most cardiovascular ischemic events. The pathophysiology of atherosclerosis involves various cell types and associated processes, including endothelial cell activation, monocyte recruitment, smooth muscle cell migration, involvement of macrophages and foam cells, and instability of the extracellular matrix. The process of endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a pivotal process in mediating vascular inflammation associated with atherosclerosis. This transition occurs gradually, with a significant portion of endothelial cells adopting an intermediate state, characterized by a partial loss of endothelial-specific gene expression and the acquisition of "mesenchymal" traits. Consequently, this shift disrupts endothelial cell junctions, increases vascular permeability, and exacerbates inflammation, creating a self-perpetuating cycle that drives atherosclerotic progression. While endothelial cell dysfunction initiates the development of atherosclerosis, autophagy, a cellular catabolic process designed to safeguard cells by recycling intracellular molecules, is believed to exert a significant role in plaque development. Identifying the pathological mechanisms and molecular mediators of EndoMT underpinning endothelial autophagy, may be of clinical relevance. Here, we offer new insights into the underlying biology of atherosclerosis and present potential molecular mechanisms of atherosclerotic resistance and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
7
|
Banerjee S, Onwunyi VRC, Hong J, Martineau S, Fishbein GA, Bonnet SB, Provencher S, Bonnet S, Umar S. RV-specific Targeting of Snai1 Rescues Pulmonary Hypertension-induced Right Ventricular Failure by Inhibiting EndMT and Fibrosis via LOXL2 Mediated Mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.30.591766. [PMID: 38746200 PMCID: PMC11092652 DOI: 10.1101/2024.04.30.591766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Pulmonary hypertension (PH)-induced right ventricular (RV) failure (PH-RVF) is a significant prognostic determinant of mortality and is characterized by RV hypertrophy, endothelial-to-mesenchymal transition (EndMT), fibroblast-to-myofibroblast transition (FMT), fibrosis, and extracellular matrix (ECM)-remodeling. Despite the importance of RV function in PH, the mechanistic details of PH-RVF, especially the regulatory control of RV EndMT, FMT, and fibrosis, remain unclear. The action of transcription factor Snai1 is shown to be mediated through LOXL2 recruitment, and their co-translocation to the nucleus, during EndMT progression. We hypothesize that RV EndMT and fibrosis in PH-RVF are governed by the TGFβ1-Snai1-LOXL2 axis. Furthermore, targeting Snai1 could serve as a novel therapeutic strategy for PH-RVF. Methods Adult male Sprague Dawley rats (250-300g) received either a single subcutaneous injection of Monocrotaline (MCT, 60mg/kg, n=9; followed for 30-days) or Sugen (SU5416 20mg/kg, n=9; 10% O 2 hypoxia for 3-weeks followed by normoxia for 2-weeks) or PBS (CTRL, n=9). We performed secondary bioinformatics analysis on the RV bulk RNA-Seq data from MCT, SuHx, and PAB rats and human PH-PVF. We validated EndMT and FMT and their association with Snai1 and LOXL2 in the RVs of MCT and SuHx rat models and human PH-RVF using immunofluorescence, qPCR, and Western blots. For in vivo Snai1 knockdown (Snai1-KD), MCT-rats either received Snai1-siRNA (n=7; 5nM/injection every 3-4 days; 4-injections) or scramble (SCRM-KD; n=7) through tail vein from day 14-30 after MCT. Echocardiography and catheterization were performed terminally. Bulk RNASeq and differential expression analysis were performed on Snai1- and SCRM-KD rat RVs. In vitro Snai1-KD was performed on human coronary artery endothelial cells (HCAECs) and human cardiac fibroblasts (HCFs) under hypoxia+TGFβ1 for 72-hrs. Results PH-RVF had increased RVSP and Fulton index and decreased RV fractional area change (RVFAC %). RV RNASeq demonstrated EndMT as the common top-upregulated pathway between rat (MCT, SuHx, and PAB) and human PH-RVF. Immunofluorescence using EndMT- and FMT-specific markers demonstrated increased EndMT and FMT in RV of MCT and SuHx rats and PH-RVF patients. Further, RV expression of TGFβ1, Snai1, and LOXL2 was increased in MCT and SuHx. Nuclear co-localization and increased immunoreactivity, transcript, and protein levels of Snai1 and LOXL2 were observed in MCT and SuHx rats and human RVs. MCT rats treated with Snai1-siRNA demonstrated decreased Snai1 expression, RVSP, Fulton index, and increased RVFAC. Snai1-KD resulted in decreased RV-EndMT, FMT, and fibrosis via a LOXL2-dependent manner. Further, Snai1-KD inhibited hypoxia+TGFβ1-induced EndMT in HCAECs and FMT in HCFs in vitro by decreasing perinuclear/nuclear Snai1+LOXL2 expression and co-localization. Conclusions RV-specific targeting of Snai1 rescues PH-RVF by inhibiting EndMT and Fibrosis via a LOXL2-mediated mechanism.
Collapse
|
8
|
Li Y, Ni SH, Liu X, Sun SN, Ling GC, Deng JP, Ou-Yang XL, Huang YS, Li H, Chen ZX, Huang XF, Xian SX, Yang ZQ, Wang LJ, Wu HY, Lu L. Crosstalk between endothelial cells with a non-canonical EndoMT phenotype and cardiomyocytes/fibroblasts via IGFBP5 aggravates TAC-induced cardiac dysfunction. Eur J Pharmacol 2024; 966:176378. [PMID: 38309679 DOI: 10.1016/j.ejphar.2024.176378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
Heart failure (HF) is a complex chronic condition characterized by structural and functional impairments. The differentiation of endothelial cells into myofibroblasts (EndoMT) in response to cardiac fibrosis is controversial, and the relative contribution of endothelial plasticity remains to be explored. Single-cell RNA sequencing was used to identify endothelial cells undergoing fibrotic differentiation within 2 weeks of transverse aortic constriction (TAC). This subset of endothelial cells transiently expressed fibrotic genes but had low expression of alpha-smooth muscle actin, indicating a non-canonical EndoMT, which we named a transient fibrotic-like phenotype (EndoFP). The role of EndoFP in pathological cardiac remodeling may be correlated with increased levels of osteopontin. Cardiomyocytes and fibroblasts co-cultured with EndoFP exhibited heightened pro-hypertrophic and pro-fibrotic effects. Mechanistically, we found that the upregulated expression of insulin-like growth factor-binding protein 5 may be a key mediator of EndoFP-induced cardiac dysfunction. Furthermore, our findings suggested that Rab5a is a novel regulatory gene involved in the EndoFP process. Our study suggests that the specific endothelial subset identified in TAC-induced pressure overload plays a critical role in the cellular interactions that lead to cardiac fibrosis and hypertrophy. Additionally, our findings provide insight into the mechanisms underlying EndoFP, making it a potential therapeutic target for early heart failure.
Collapse
Affiliation(s)
- Yue Li
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, 518000, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Shi-Hao Ni
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Xin Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shu-Ning Sun
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Gui-Chen Ling
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jian-Ping Deng
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Xiao-Lu Ou-Yang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Yu-Sheng Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Huan Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Zi-Xin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Xiu-Fang Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Shao-Xiang Xian
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Zhong-Qi Yang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Ling-Jun Wang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China.
| | - Hong-Yan Wu
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, 518000, China.
| | - Lu Lu
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China.
| |
Collapse
|
9
|
Bréchot N, Rutault A, Marangon I, Germain S. Blood endothelium transition and phenotypic plasticity: A key regulator of integrity/permeability in response to ischemia. Semin Cell Dev Biol 2024; 155:16-22. [PMID: 37479554 DOI: 10.1016/j.semcdb.2023.07.004] [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: 04/20/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
In the human body, the 1013 blood endothelial cells (ECs) which cover a surface of 500-700 m2 (Mai et al., 2013) are key players of tissue homeostasis, remodeling and regeneration. Blood vessel ECs play a major role in the regulation of metabolic and gaz exchanges, cell trafficking, blood coagulation, vascular tone, blood flow and fluid extravasation (also referred to as blood vascular permeability). ECs are heterogeneous in various capillary beds and have the exquisite capacity to cope with environmental changes by regulating their gene expression. Ischemia has major detrimental effects on the endothelium and ischemia-induced regulation of vascular integrity is of paramount importance for human health, as small amounts of fluid accumulation in the interstitium may be responsible for major effects on organ functions and patients outcome. In this review, we will here focus on the stimuli and the molecular mechanisms that control blood endothelium maintenance and phenotypic plasticity/transition involved in controlling blood capillary leakage that might open new avenues for therapeutic applications.
Collapse
Affiliation(s)
- Nicolas Bréchot
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France; Intensive Care Medicine Department, Université de Paris Cité, Hôpital européen Georges-Pompidou, AP-HP, AP-HP.CUP, 75015 Paris, France.
| | - Alexandre Rutault
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Iris Marangon
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France.
| |
Collapse
|
10
|
Takahashi K, Kobayashi M, Katsumata H, Tokizaki S, Anzai T, Ikeda Y, Alcaide DM, Maeda K, Ishihara M, Tahara K, Kubota Y, Itoh F, Park J, Takahashi K, Matsunaga YT, Yoshimatsu Y, Podyma‐Inoue KA, Watabe T. CD40 is expressed in the subsets of endothelial cells undergoing partial endothelial-mesenchymal transition in tumor microenvironment. Cancer Sci 2024; 115:490-506. [PMID: 38111334 PMCID: PMC10859613 DOI: 10.1111/cas.16045] [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: 07/27/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/20/2023] Open
Abstract
Tumor progression and metastasis are regulated by endothelial cells undergoing endothelial-mesenchymal transition (EndoMT), a cellular differentiation process in which endothelial cells lose their properties and differentiate into mesenchymal cells. The cells undergoing EndoMT differentiate through a spectrum of intermediate phases, suggesting that some cells remain in a partial EndoMT state and exhibit an endothelial/mesenchymal phenotype. However, detailed analysis of partial EndoMT has been hampered by the lack of specific markers. Transforming growth factor-β (TGF-β) plays a central role in the induction of EndoMT. Here, we showed that inhibition of TGF-β signaling suppressed EndoMT in a human oral cancer cell xenograft mouse model. By using genetic labeling of endothelial cell lineage, we also established a novel EndoMT reporter cell system, the EndoMT reporter endothelial cells (EMRECs), which allow visualization of sequential changes during TGF-β-induced EndoMT. Using EMRECs, we characterized the gene profiles of multiple EndoMT stages and identified CD40 as a novel partial EndoMT-specific marker. CD40 expression was upregulated in the cells undergoing partial EndoMT, but decreased in the full EndoMT cells. Furthermore, single-cell RNA sequencing analysis of human tumors revealed that CD40 expression was enriched in the population of cells expressing both endothelial and mesenchymal cell markers. Moreover, decreased expression of CD40 in EMRECs enhanced TGF-β-induced EndoMT, suggesting that CD40 expressed during partial EndoMT inhibits transition to full EndoMT. The present findings provide a better understanding of the mechanisms underlying TGF-β-induced EndoMT and will facilitate the development of novel therapeutic strategies targeting EndoMT-driven cancer progression and metastasis.
Collapse
Affiliation(s)
- Kazuki Takahashi
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Institute of Industrial ScienceThe University of TokyoTokyoJapan
| | - Miho Kobayashi
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Hisae Katsumata
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Shiori Tokizaki
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Department of Oral and Maxillofacial Surgical Oncology, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Tatsuhiko Anzai
- Department of Biostatistics, M&D Data Science CenterTokyo Medical and Dental UniversityTokyoJapan
| | - Yukinori Ikeda
- Institute of Industrial ScienceThe University of TokyoTokyoJapan
| | | | - Kentaro Maeda
- Laboratory of Oncology, School of Life SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Makoto Ishihara
- Scientific Affairs Section, Life Science Sales Department, Life Science Business Division, Medical Business GroupSony CorporationKanagawaJapan
| | - Katsutoshi Tahara
- Section 1, Product Design Department 2, Medical Product Design Division, Medical Business GroupSony CorporationKanagawaJapan
| | - Yoshiaki Kubota
- Department of AnatomyKeio University School of MedicineTokyoJapan
| | - Fumiko Itoh
- Laboratory of Stem Cells RegulationsTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Jihwan Park
- School of Life SciencesGwangju Institute of Science and Technology (GIST)GwangjuSouth Korea
| | - Kunihiko Takahashi
- Department of Biostatistics, M&D Data Science CenterTokyo Medical and Dental UniversityTokyoJapan
| | | | - Yasuhiro Yoshimatsu
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Laboratory of Oncology, School of Life SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
- Division of Pharmacology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Katarzyna A. Podyma‐Inoue
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
| | - Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental SciencesTokyo Medical and Dental UniversityTokyoJapan
- Laboratory of Oncology, School of Life SciencesTokyo University of Pharmacy and Life SciencesTokyoJapan
| |
Collapse
|
11
|
Hussein MA, Valinezhad K, Adel E, Munirathinam G. MALAT-1 Is a Key Regulator of Epithelial-Mesenchymal Transition in Cancer: A Potential Therapeutic Target for Metastasis. Cancers (Basel) 2024; 16:234. [PMID: 38201661 PMCID: PMC10778055 DOI: 10.3390/cancers16010234] [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: 12/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Metastasis-associated lung adenocarcinoma transcript-1 (MALAT-1) is a long intergenic non-coding RNA (lncRNA) located on chr11q13. It is overexpressed in several cancers and controls gene expression through chromatin modification, transcriptional regulation, and post-transcriptional regulation. Importantly, MALAT-1 stimulates cell proliferation, migration, and metastasis and serves a vital role in driving the epithelial-to-mesenchymal transition (EMT), subsequently acquiring cancer stem cell-like properties and developing drug resistance. MALAT-1 modulates EMT by interacting with various intracellular signaling pathways, notably the phosphoinositide 3-kinase (PI3K)/Akt and Wnt/β-catenin pathways. It also behaves like a sponge for microRNAs, preventing their interaction with target genes and promoting EMT. In addition, we have used bioinformatics online tools to highlight the disparities in the expression of MALAT-1 between normal and cancer samples using data from The Cancer Genome Atlas (TCGA). Furthermore, the intricate interplay of MALAT-1 with several essential targets of cancer progression and metastasis renders it a good candidate for therapeutic interventions. Several innovative approaches have been exploited to target MALAT-1, such as short hairpin RNAs (shRNAs), antisense oligonucleotides (ASOs), and natural products. This review emphasizes the interplay between MALAT-1 and EMT in modulating cancer metastasis, stemness, and chemoresistance in different cancers.
Collapse
Affiliation(s)
- Mohamed Ali Hussein
- Department of Pharmaceutical Services, Children’s Cancer Hospital Egypt, Cairo 57357, Egypt;
- Department of Biology, School of Sciences and Engineering, American University in Cairo, New Cairo 11835, Egypt;
| | - Kamyab Valinezhad
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL 61107, USA;
| | - Eman Adel
- Department of Biology, School of Sciences and Engineering, American University in Cairo, New Cairo 11835, Egypt;
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, College of Medicine, University of Illinois, Rockford, IL 61107, USA;
| |
Collapse
|
12
|
Jimenez SA, Piera-Velazquez S. Cellular Transdifferentiation: A Crucial Mechanism of Fibrosis in Systemic Sclerosis. Curr Rheumatol Rev 2024; 20:388-404. [PMID: 37921216 DOI: 10.2174/0115733971261932231025045400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 11/04/2023]
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology with a highly complex pathogenesis that despite extensive investigation is not completely understood. The clinical and pathologic manifestations of the disease result from three distinct processes: 1) Severe and frequently progressive tissue fibrosis causing exaggerated and deleterious accumulation of interstitial collagens and other extracellular matrix molecules in the skin and various internal organs; 2) extensive fibroproliferative vascular lesions affecting small arteries and arterioles causing tissue ischemic alterations; and 3) cellular and humoral immunity abnormalities with the production of numerous autoantibodies, some with very high specificity for SSc. The fibrotic process in SSc is one of the main causes of disability and high mortality of the disease. Owing to its essentially universal presence and the severity of its clinical effects, the mechanisms involved in the development and progression of tissue fibrosis have been extensively investigated, however, despite intensive investigation, the precise molecular mechanisms have not been fully elucidated. Several recent studies have suggested that cellular transdifferentiation resulting in the phenotypic conversion of various cell types into activated myofibroblasts may be one important mechanism. Here, we review the potential role that cellular transdifferentiation may play in the development of severe and often progressive tissue fibrosis in SSc.
Collapse
Affiliation(s)
- Sergio A Jimenez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| | - Sonsoles Piera-Velazquez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| |
Collapse
|
13
|
Zhao Y, Xiong W, Li C, Zhao R, Lu H, Song S, Zhou Y, Hu Y, Shi B, Ge J. Hypoxia-induced signaling in the cardiovascular system: pathogenesis and therapeutic targets. Signal Transduct Target Ther 2023; 8:431. [PMID: 37981648 PMCID: PMC10658171 DOI: 10.1038/s41392-023-01652-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/10/2023] [Accepted: 09/13/2023] [Indexed: 11/21/2023] Open
Abstract
Hypoxia, characterized by reduced oxygen concentration, is a significant stressor that affects the survival of aerobic species and plays a prominent role in cardiovascular diseases. From the research history and milestone events related to hypoxia in cardiovascular development and diseases, The "hypoxia-inducible factors (HIFs) switch" can be observed from both temporal and spatial perspectives, encompassing the occurrence and progression of hypoxia (gradual decline in oxygen concentration), the acute and chronic manifestations of hypoxia, and the geographical characteristics of hypoxia (natural selection at high altitudes). Furthermore, hypoxia signaling pathways are associated with natural rhythms, such as diurnal and hibernation processes. In addition to innate factors and natural selection, it has been found that epigenetics, as a postnatal factor, profoundly influences the hypoxic response and progression within the cardiovascular system. Within this intricate process, interactions between different tissues and organs within the cardiovascular system and other systems in the context of hypoxia signaling pathways have been established. Thus, it is the time to summarize and to construct a multi-level regulatory framework of hypoxia signaling and mechanisms in cardiovascular diseases for developing more therapeutic targets and make reasonable advancements in clinical research, including FDA-approved drugs and ongoing clinical trials, to guide future clinical practice in the field of hypoxia signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Yongchao Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Weidong Xiong
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Shuai Song
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - You Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yiqing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Junbo Ge
- Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, 200032, China.
- Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Chen WC, Yu WK, Su VYF, Hsu HS, Yang KY. NLRP3 Inflammasome Activates Endothelial-to-Mesenchymal Transition via Focal Adhesion Kinase Pathway in Bleomycin-Induced Pulmonary Fibrosis. Int J Mol Sci 2023; 24:15813. [PMID: 37958797 PMCID: PMC10648980 DOI: 10.3390/ijms242115813] [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: 09/17/2023] [Revised: 10/07/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Idiopathic pulmonary fibrosis has poor clinical outcomes despite antifibrotic treatment. The nucleotide-binding domain leucine-rich repeat-containing receptor, pyrin domain-containing-3 (NLRP3) inflammasome and endothelial-to-mesenchymal transition (EndoMT) were shown to be involved in the pathogenesis of pulmonary fibrosis. However, the detailed mechanism is unknown. Our study aimed to investigate the role of the NLRP3 inflammasome in the regulation of EndoMT in pulmonary fibrosis. The inhibition of the NLRP3 inflammasome via a caspase-1 inhibitor, Ac-YVAD-cmk (YVAD), was intraperitoneally administered to male C57BL/6 mice (8-12 weeks old) one hour before bleomycin intratracheal injection (1.5 U/kg). Immunohistochemical staining, Masson's trichrome staining, enzyme-linked immunosorbent assay, immunofluorescence, and Western blotting were used to assess the activity of the NLRP3 inflammasome and EndoMT in lung samples from mice. Human pulmonary microvascular endothelial cells (HPMECs) were used as a model of EndoMT in vitro with YVAD and bleomycin stimulation. We observed the activation of the NLRP3 inflammasome and EndoMT (decreased vascular endothelial cadherin with increased alpha-smooth muscle actin and vimentin) in the lung samples after bleomycin. However, inhibition of the NLRP3 inflammasome significantly reduces EndoMT via inhibiting focal adhesion kinase (FAK). In vitro studies also confirmed these findings. In conclusion, NLRP3 inflammasome inhibition could reduce lung inflammation and fibrosis via the regulation of EndoMT by the FAK pathway.
Collapse
Affiliation(s)
- Wei-Chih Chen
- Institute of Emergency and Critical Care Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-C.C.); (H.-S.H.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-K.Y.); (V.Y.-F.S.)
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Wen-Kuang Yu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-K.Y.); (V.Y.-F.S.)
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Institute of Physiology, College of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Vincent Yi-Fong Su
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-K.Y.); (V.Y.-F.S.)
- Department of Internal Medicine, Taipei City Hospital, Taipei 110, Taiwan
| | - Han-Shui Hsu
- Institute of Emergency and Critical Care Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-C.C.); (H.-S.H.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-K.Y.); (V.Y.-F.S.)
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Kuang-Yao Yang
- Institute of Emergency and Critical Care Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-C.C.); (H.-S.H.)
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (W.-K.Y.); (V.Y.-F.S.)
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Cancer Progression Research Center, National Yang-Ming University, Taipei 112, Taiwan
| |
Collapse
|
15
|
Nijim W, Moustafa M, Humble J, Al-Shabrawey M. Endothelial to mesenchymal cell transition in diabetic retinopathy: targets and therapeutics. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1230581. [PMID: 38983088 PMCID: PMC11182279 DOI: 10.3389/fopht.2023.1230581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/11/2023] [Indexed: 07/11/2024]
Abstract
Diabetic retinopathy (DR) is a result of neurovacular insults from hyperglycemia in diabetes mellitus (DM), and it is one of the top causes of vision loss throughout the modern world. This review article explores the role endothelial to mesenchymal transition (EndMT) has on the pathogenesis of DR. EndMT contributes to the disruption of the blood-retinal barrier, vascular leakage, neovascularization, and fibrosis observed in DR. Risk factors and biomarkers associated with DR severity are discussed, highlighting the importance of early detection and targeted therapies. Current treatments primarily focus on anti-vascular endothelial growth factor (anti-VEGF) agents, corticosteroids, and laser photocoagulation. However, emerging therapeutic strategies aimed at inhibiting EndMT and its downstream effects show promise in preventing the development and progression of DR. Understanding the molecular and cellular mechanisms underlying EndMT in DR provides valuable insights into the disease process and offers potential options for the development of potential treatments.
Collapse
Affiliation(s)
- Wasef Nijim
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mohamed Moustafa
- Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
- Eye Research Institute, Oakland University, Rochester, MI, United States
| | - Julia Humble
- Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
- Eye Research Institute, Oakland University, Rochester, MI, United States
| | - Mohamed Al-Shabrawey
- Eye Research Center, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
- Eye Research Institute, Oakland University, Rochester, MI, United States
- Foundation Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, United States
| |
Collapse
|
16
|
Davies A, Zoubeidi A, Beltran H, Selth LA. The Transcriptional and Epigenetic Landscape of Cancer Cell Lineage Plasticity. Cancer Discov 2023; 13:1771-1788. [PMID: 37470668 PMCID: PMC10527883 DOI: 10.1158/2159-8290.cd-23-0225] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/25/2023] [Accepted: 06/09/2023] [Indexed: 07/21/2023]
Abstract
Lineage plasticity, a process whereby cells change their phenotype to take on a different molecular and/or histologic identity, is a key driver of cancer progression and therapy resistance. Although underlying genetic changes within the tumor can enhance lineage plasticity, it is predominantly a dynamic process controlled by transcriptional and epigenetic dysregulation. This review explores the transcriptional and epigenetic regulators of lineage plasticity and their interplay with other features of malignancy, such as dysregulated metabolism, the tumor microenvironment, and immune evasion. We also discuss strategies for the detection and treatment of highly plastic tumors. SIGNIFICANCE Lineage plasticity is a hallmark of cancer and a critical facilitator of other oncogenic features such as metastasis, therapy resistance, dysregulated metabolism, and immune evasion. It is essential that the molecular mechanisms of lineage plasticity are elucidated to enable the development of strategies to effectively target this phenomenon. In this review, we describe key transcriptional and epigenetic regulators of cancer cell plasticity, in the process highlighting therapeutic approaches that may be harnessed for patient benefit.
Collapse
Affiliation(s)
- Alastair Davies
- Oncology Research Discovery, Pfizer Worldwide Research and Development, San Diego, CA, USA
| | - Amina Zoubeidi
- Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Vancouver Prostate Centre, Vancouver, British Columbia, Canada
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Luke A. Selth
- Flinders Health and Medical Research Institute and Freemasons Centre for Male Health and Wellbeing, Flinders University, Bedford Park, South Australia, 5042 Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5005 Australia
| |
Collapse
|
17
|
Bouhamida E, Morciano G, Pedriali G, Ramaccini D, Tremoli E, Giorgi C, Pinton P, Patergnani S. The Complex Relationship between Hypoxia Signaling, Mitochondrial Dysfunction and Inflammation in Calcific Aortic Valve Disease: Insights from the Molecular Mechanisms to Therapeutic Approaches. Int J Mol Sci 2023; 24:11105. [PMID: 37446282 DOI: 10.3390/ijms241311105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS) is among the most common causes of cardiovascular mortality in an aging population worldwide. The pathomechanisms of CAVS are such a complex and multifactorial process that researchers are still making progress to understand its physiopathology as well as the complex players involved in CAVS pathogenesis. Currently, there is no successful and effective treatment to prevent or slow down the disease. Surgical and transcatheter valve replacement represents the only option available for treating CAVS. Insufficient oxygen availability (hypoxia) has a critical role in the pathogenesis of almost all CVDs. This process is orchestrated by the hallmark transcription factor, hypoxia-inducible factor 1 alpha subunit (HIF-1α), which plays a pivotal role in regulating various target hypoxic genes and metabolic adaptations. Recent studies have shown a great deal of interest in understanding the contribution of HIF-1α in the pathogenesis of CAVS. However, it is deeply intertwined with other major contributors, including sustained inflammation and mitochondrial impairments, which are attributed primarily to CAVS. The present review aims to cover the latest understanding of the complex interplay effect of hypoxia signaling pathways, mitochondrial dysfunction, and inflammation in CAVS. We propose further hypotheses and interconnections on the complexity of these impacts in a perspective of better understanding the pathophysiology. These interplays will be examined considering recent studies that shall help us better dissect the molecular mechanism to enable the design and development of potential future therapeutic approaches that can prevent or slow down CAVS processes.
Collapse
Affiliation(s)
- Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
18
|
Strelnikova EA, Kalinin RE, Suchkov IA, Korotkova NV, Mzhavanadze ND. [Molecular and Cellular Aspects of the Endothelial-Mesenchymal Transition in Cardiovascular Diseases]. Mol Biol 2023; 57:563-572. [PMID: 37528777 DOI: 10.1134/s0026893323030111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/30/2022] [Accepted: 10/30/2022] [Indexed: 10/10/2024]
Abstract
Endothelial cells (ECs), which form the inner surface of the blood vessels, contact the blood, withstand mechanical pressure, and demonstrate heterogeneous reactions to exogenous and endogenous stimuli. ECs have unique properties in accordance with their niches and play an important role in regulating vascular homeostasis. Endothelial cells may undergo a dynamic phenotypic switch in terms of its heterogeneity, which may lead to endothelial dysfunction and a number of associated pathologies. Endothelial-mesenchymal transition (EndMT) is one of the possible molecular and cellular mechanisms of this kind. EndMT is characterized by phenotypic changes in ECs through which endothelial cells acquire new properties, i.e., start producing mesenchymal markers such as alpha-SMA and vimentin, change morphology, and become able to migrate. EndMT is a complex biological process that can be induced by inflammation, hypoxia, or oxidative stress and be involved in pathogenesis of cardiovascular disease. This review describes the key markers, inhibitors, and inducers of endothelial-mesenchymal transition and overall state-of-the-art of EndMT in cardiovascular diseases.
Collapse
Affiliation(s)
- E A Strelnikova
- Pavlov Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, 390026 Russia
| | - R E Kalinin
- Pavlov Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, 390026 Russia
| | - I A Suchkov
- Pavlov Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, 390026 Russia
| | - N V Korotkova
- Pavlov Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, 390026 Russia
| | - N D Mzhavanadze
- Pavlov Ryazan State Medical University, Ministry of Health of the Russian Federation, Ryazan, 390026 Russia
| |
Collapse
|
19
|
Wiktorska M, Sacewicz-Hofman I, Niewiarowska J. The endothelial-to-mesenchymal transition changes the focal adhesion site proteins levels and the SLRP-lumican level in HMEC-1 cell line. Exp Cell Res 2023:113692. [PMID: 37392962 DOI: 10.1016/j.yexcr.2023.113692] [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: 01/25/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Scleroderma, the chronic autoimmune disease is a consequence of inflammation in the connective tissue. Prolonged duration affects formation of compact connective tissue strands (scarring) within the target organ. Endothelial cells undergoing endothelial-to-mesenchymal transition (EndMT) are the source of fibroblast phenotype-resembling cells. EndMT contributes to reorganization of the focal adhesion proteins (FA), including integrins, and intensive extracellular matrix (ECM) remodelling. However, in endothelial cells, the relationship between EndMT and the interaction of integrin receptors with lumican - a component of ECM, is still unclear. Our findings indicate that at the early stages of EndMT caused by Snail-1 transcription factor overexpression, the level of the β1 integrin subunit and its phosphorylation are elevated. Simultaneously, the changes in the level of proteins that build FAs and promote activation of integrin receptors as well as a decrease in lumican quantity were observed. These modulations contributed to increased migration of human microvascular endothelial cells, HMEC-1. Our findings were achieved by WB, ELISA and wound healing assay. Taken altogether, transfection of HMEC-1 cells with Snail-1 plasmids inducing the early stages of EndMT results in the increase of total FAK and integrin β1 phosphorylation as well as cell migration: phenomena which are modulated by interaction with lumican.
Collapse
Affiliation(s)
- Magdalena Wiktorska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, 92-215, Lodz, Poland
| | | | - Jolanta Niewiarowska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, 92-215, Lodz, Poland.
| |
Collapse
|
20
|
Kalinin R, Suchkov I, Mzhavanadze N, Surov I. The role of endothelial-mesenchymal transition in vascular restenosis after endovascular treatment in patients with peripheral arterial disease of the lower extremities. RUSSIAN JOURNAL OF CARDIOLOGY AND CARDIOVASCULAR SURGERY 2023; 16:469. [DOI: 10.17116/kardio202316051469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Symptomatic atherosclerosis of the lower extremity arteries is becoming more prevalent each year. Atherosclerotic stenosis or occlusions of femoral and popliteal arteries may often co-exist with aorto-iliac or distal lesions. Treatment options depend on severity of disease, localization of atherosclerotic plaques and comorbidities. The main objective of treating patients with atherosclerotic peripheral artery disease affecting lower extremities is limb salvage and relief of symptoms. Endovascular interventions are able to improve limb salve rates and prognosis of disease, as well as to avoid open and often traumatic reconstructive surgical procedures on the lower extremities. However, postoperative restenosis is still one of the key problems following percutaneous angioplasty and/or stenting. Endothelial mesenchymal transition is regarded as a possible key pathogenetic mechanism underlying restenosis. This article is dedicated to EndMT and its possible role in restenosis.
Collapse
|
21
|
Watabe T, Takahashi K, Pietras K, Yoshimatsu Y. Roles of TGF-β signals in tumor microenvironment via regulation of the formation and plasticity of vascular system. Semin Cancer Biol 2023; 92:130-138. [PMID: 37068553 DOI: 10.1016/j.semcancer.2023.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 04/19/2023]
Abstract
Tumor cells evolve in tumor microenvironment composed of multiple cell types. Among these, endothelial cells (ECs) are the major players in tumor angiogenesis, which is a driver of tumor progression and metastasis. Increasing evidence suggests that ECs also contribute to tumor progression and metastasis as they modify their phenotypes to differentiate into mesenchymal cells through a process known as endothelial-mesenchymal transition (EndoMT). This plasticity of ECs is mediated by various cytokines, including transforming growth factor-β (TGF-β), and modulated by other stimuli depending on the cellular contexts. Recent lines of evidence have shown that EndoMT is involved in various steps of tumor progression, including tumor angiogenesis, intravasation and extravasation of cancer cells, formation of cancer-associated fibroblasts, and cancer therapy resistance. In this review, we summarize current updates on EndoMT, highlight the roles of EndoMT in tumor progression and metastasis, and underline targeting EndoMT as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Tetsuro Watabe
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| | - Kazuki Takahashi
- Department of Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Institute of Industrial Science, The University of Tokyo, Tokyo, Japan.
| | - Kristian Pietras
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University Cancer Centre, Medicon Village, Lund University, 223 81 Lund, Sweden.
| | - Yasuhiro Yoshimatsu
- Division of Pharmacology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| |
Collapse
|
22
|
Yang L, Wan N, Gong F, Wang X, Feng L, Liu G. Transcription factors and potential therapeutic targets for pulmonary hypertension. Front Cell Dev Biol 2023; 11:1132060. [PMID: 37009479 PMCID: PMC10064017 DOI: 10.3389/fcell.2023.1132060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/03/2023] [Indexed: 03/19/2023] Open
Abstract
Pulmonary hypertension (PH) is a refractory and fatal disease characterized by excessive pulmonary arterial cell remodeling. Uncontrolled proliferation and hypertrophy of pulmonary arterial smooth muscle cells (PASMCs), dysfunction of pulmonary arterial endothelial cells (PAECs), and abnormal perivascular infiltration of immune cells result in pulmonary arterial remodeling, followed by increased pulmonary vascular resistance and pulmonary pressure. Although various drugs targeting nitric oxide, endothelin-1 and prostacyclin pathways have been used in clinical settings, the mortality of pulmonary hypertension remains high. Multiple molecular abnormalities have been implicated in pulmonary hypertension, changes in numerous transcription factors have been identified as key regulators in pulmonary hypertension, and a role for pulmonary vascular remodeling has been highlighted. This review consolidates evidence linking transcription factors and their molecular mechanisms, from pulmonary vascular intima PAECs, vascular media PASMCs, and pulmonary arterial adventitia fibroblasts to pulmonary inflammatory cells. These findings will improve the understanding of particularly interactions between transcription factor-mediated cellular signaling pathways and identify novel therapies for pulmonary hypertension.
Collapse
Affiliation(s)
- Liu Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Naifu Wan
- Department of Vascular & Cardiology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fanpeng Gong
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xianfeng Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Guizhu Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- *Correspondence: Guizhu Liu,
| |
Collapse
|
23
|
Abstract
The endothelium is one of the largest organ systems in the body, and data continue to emerge regarding the importance of endothelial cell (EC) dysfunction in vascular aging and a range of cardiovascular diseases (CVDs). Over the last two decades and as a process intimately related to EC dysfunction, an increasing number of studies have also implicated endothelial to mesenchymal transition (EndMT) as a potentially disease-causal pathobiologic process that is involved in a multitude of differing CVDs. However, EndMT is also involved in physiologic processes (e.g., cardiac development), and transient EndMT may contribute to vascular regeneration in certain contexts. Given that EndMT involves a major alteration in the EC-specific molecular program, and that it potentially contributes to CVD pathobiology, the clinical translation opportunities are significant, but further molecular and translational research is needed to see these opportunities realized.
Collapse
Affiliation(s)
- Yang Xu
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; .,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
24
|
Fan M, Yang K, Wang X, Chen L, Gill PS, Ha T, Liu L, Lewis NH, Williams DL, Li C. Lactate promotes endothelial-to-mesenchymal transition via Snail1 lactylation after myocardial infarction. SCIENCE ADVANCES 2023; 9:eadc9465. [PMID: 36735787 PMCID: PMC9897666 DOI: 10.1126/sciadv.adc9465] [Citation(s) in RCA: 163] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 01/03/2023] [Indexed: 06/01/2023]
Abstract
High levels of lactate are positively associated with the prognosis and mortality in patients with heart attack. Endothelial-to-mesenchymal transition (EndoMT) plays an important role in cardiac fibrosis. Here, we report that lactate exerts a previously unknown function that increases cardiac fibrosis and exacerbates cardiac dysfunction by promoting EndoMT following myocardial infarction (MI). Treatment of endothelial cells with lactate disrupts endothelial cell function and induces mesenchymal-like function following hypoxia by activating the TGF-β/Smad2 pathway. Mechanistically, lactate induces an association between CBP/p300 and Snail1, leading to lactylation of Snail1, a TGF-β transcription factor, through lactate transporter monocarboxylate transporter (MCT)-dependent signaling. Inhibiting Snail1 diminishes lactate-induced EndoMT and TGF-β/Smad2 activation after hypoxia/MI. The MCT inhibitor CHC mitigates lactate-induced EndoMT and Snail1 lactylation. Silence of MCT1 compromises lactate-promoted cardiac dysfunction and EndoMT after MI. We conclude that lactate acts as an important molecule that up-regulates cardiac EndoMT after MI via induction of Snail1 lactylation.
Collapse
Affiliation(s)
- Min Fan
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Kun Yang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Xiaohui Wang
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Linjian Chen
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, China
| | - P. Spencer Gill
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Tuanzhu Ha
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Li Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nicole H. Lewis
- Department of Medical Education, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - David L. Williams
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Chuanfu Li
- Department of Surgery, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
- The Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
25
|
The Carthamus tinctorius L. and Lepidium apetalum Willd. Drug Pair Inhibits EndMT through the TGF β1/Snail Signaling Pathway in the Treatment of Myocardial Fibrosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:6018375. [PMID: 36686974 PMCID: PMC9851799 DOI: 10.1155/2023/6018375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023]
Abstract
Background Myocardial fibrosis (MF) is an essential pathological factor for heart failure. Previous studies have shown that the combination of Carthamus tinctorius L. and Lepidium apetalum Willd. (C-L), two types of Chinese herbal medicine, can ameliorate MF after myocardial infarction (MI) in rats and inhibit the activation of myocardial fibroblasts. However, the mechanism of C-L in the treatment of MF remains unclear. Methods A rat model of MF with left anterior descending coronary ligation-induced MI was first established. Then, the effects of C-L on cardiac function, MF, and endothelial-to-mesenchymal transition (EndMT) were evaluated by the left ventricular ejection fraction (LVEF), serum N-terminal pro-brain natriuretic peptide (NT-proBNP) levels, Masson's trichrome staining, and immunohistochemical and immunofluorescence staining. Next, a hypoxia-induced cardiac microvascular endothelial cell (CMEC) model was established to observe the effects of C-L on EndMT. The supernatant of CMECs was collected and used to culture cardiac fibroblasts (CFs) and observe the effects of CMEC paracrine factors on CFs. Results Animal experiments indicated that C-L improves the cardiac function of rats after MI, inhibits the progression of EndMT and MF, and downregulates TGFβ1, Snail, and CTGF expression. Cell experiments showed that drug-loaded serum containing C-L inhibits the EndMT of CMECs under hypoxic conditions. The culture supernatant of CMECs grown under hypoxic conditions significantly activated CFs. After treatment with C-L, the activating factor for CFs in hypoxic CMEC culture supernatant was substantially downregulated, and the effect of the culture supernatant on CF activation was also reduced. However, TGFβ1 agonists inhibited the effects of C-L on CMECs and CFs. Conclusion Our data demonstrated that by regulating the TGFβ1/Snail pathway, C-L inhibits EndMT of CMECs and reduces the release of CF-activating factors in cells undergoing EndMT.
Collapse
|
26
|
Körbelin J, Klein J, Matuszcak C, Runge J, Harbaum L, Klose H, Hennigs JK. Transcription factors in the pathogenesis of pulmonary arterial hypertension-Current knowledge and therapeutic potential. Front Cardiovasc Med 2023; 9:1036096. [PMID: 36684555 PMCID: PMC9853303 DOI: 10.3389/fcvm.2022.1036096] [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: 09/03/2022] [Accepted: 11/21/2022] [Indexed: 01/09/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by elevated pulmonary vascular resistance and pulmonary artery pressure. Mortality remains high in severe cases despite significant advances in management and pharmacotherapy. Since currently approved PAH therapies are unable to significantly reverse pathological vessel remodeling, novel disease-modifying, targeted therapeutics are needed. Pathogenetically, PAH is characterized by vessel wall cell dysfunction with consecutive remodeling of the pulmonary vasculature and the right heart. Transcription factors (TFs) regulate the process of transcribing DNA into RNA and, in the pulmonary circulation, control the response of pulmonary vascular cells to macro- and microenvironmental stimuli. Often, TFs form complex protein interaction networks with other TFs or co-factors to allow for fine-tuning of gene expression. Therefore, identification of the underlying molecular mechanisms of TF (dys-)function is essential to develop tailored modulation strategies in PAH. This current review provides a compendium-style overview of TFs and TF complexes associated with PAH pathogenesis and highlights their potential as targets for vasculoregenerative or reverse remodeling therapies.
Collapse
Affiliation(s)
- Jakob Körbelin
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,*Correspondence: Jakob Körbelin,
| | - Julius Klein
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christiane Matuszcak
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johannes Runge
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Harbaum
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans Klose
- Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan K. Hennigs
- ENDomics Lab, Department of Medicine, Center of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Division of Pneumology and Center for Pulmonary Arterial Hypertension Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany,Jan K. Hennigs,
| |
Collapse
|
27
|
Wang E, Wang H, Chakrabarti S. Endothelial-to-mesenchymal transition: An underappreciated mediator of diabetic complications. Front Endocrinol (Lausanne) 2023; 14:1050540. [PMID: 36777351 PMCID: PMC9911675 DOI: 10.3389/fendo.2023.1050540] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Diabetes and its complications represent a great burden on the global healthcare system. Diabetic complications are fundamentally diseases of the vasculature, with endothelial cells being the centerpiece of early hyperglycemia-induced changes. Endothelial-to-mesenchymal transition is a tightly regulated process that results in endothelial cells losing endothelial characteristics and developing mesenchymal traits. Although endothelial-to-mesenchymal transition has been found to occur within most of the major complications of diabetes, it has not been a major focus of study or a common target in the treatment or prevention of diabetic complications. In this review we summarize the importance of endothelial-to-mesenchymal transition in each major diabetic complication, examine specific mechanisms at play, and highlight potential mechanisms to prevent endothelial-to-mesenchymal transition in each of the major chronic complications of diabetes.
Collapse
|
28
|
Li ZX, Chen JX, Zheng ZJ, Cai WJ, Yang XB, Huang YY, Gong Y, Xu F, Chen YS, Lin L. TGF-β1 promotes human breast cancer angiogenesis and malignant behavior by regulating endothelial-mesenchymal transition. Front Oncol 2022; 12:1051148. [PMID: 36465358 PMCID: PMC9709251 DOI: 10.3389/fonc.2022.1051148] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/18/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Endothelial-mesenchymal transition (EndMT) is an important process of angiogenesis, which plays a significant role in in tumor invasion and metastasis, while its regulatory mechanisms in breast cancer remain to be fully elucidated. We previously demonstrated that tumor-associated macrophages (TAMs) can induce EndMT in endothelial cells by secreting CCL18 through the activation of the TGF-β and Notch signaling pathways in breast cancer. This study was designed to study the role of EndMT in breast cancer angiogenesis and progression in order to explore the underlying mechanism. METHODS Immunohistochemistry (IHC) was used to evaluate the expression of microvascular density (MVD) and EndMT markers in breast cancer. TGF-β1 was used to induce EndMT models of differentiated-endothelial breast cancer stem-like cells (BCSLCs). In vitro cell migration, proliferation and matrigel tube-formation assays, as well as in vivo nude mouse tumor-bearing model and nude mouse dorsal skinfold window chamber (DSWC) model, were utilized to investigate the effects in order to explore the mechanism of EndMT induced by TGF-β1 on breast cancer progression. RESULTS In this study, we demonstrated that the EndMT markers were positively associated with MVD indicating unfavorable prognosis of invasive ductal carcinoma (IDC) patients. Functionally, TGF-β1 promoted migration, proliferation and angiogenesis of differentiated-endothelial BCSLCs by inducing EndMT in vitro and promoted tumor growth and angiogenesis in vivo. Mechanically, we revealed TGF-β1 induced EndMT by activation of TGF-β and Notch signaling pathways with increase of p-Smad2/3 and Notch1 expression. Moreover, we found Snail and Slug were key factors of TGF-β and Notch signaling pathways. CONCLUSION Our findings elucidated the mechanism of TGF-β1 in the promotion of angiogenesis and progression by EndMT in breast cancer.
Collapse
Affiliation(s)
- Zi-Xiong Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Jie-Xin Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Ze-Jun Zheng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wang-Jing Cai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Xiong-Bin Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yuan-Yuan Huang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yao Gong
- Department of Rheumatology, Shantou University Medical College, Shantou, China
| | - Feng Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yong-Song Chen
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Ling Lin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Department of Rheumatology, Shantou University Medical College, Shantou, China
| |
Collapse
|
29
|
Ro YT, Patterson JL. Transcriptional induction of TGF-β1 and endothelial-to-mesenchymal transition cell markers in human umbilical vein endothelial cells by Ebola virus infection. Genes Genomics 2022; 44:1499-1507. [DOI: 10.1007/s13258-022-01333-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/16/2022] [Indexed: 11/06/2022]
|
30
|
Liu J, Qiu R, Liu R, Song P, Lin P, Chen H, Zhou D, Wang A, Jin Y. YPEL3 Negatively Regulates Endometrial Function via the Wnt/β-Catenin Pathways during Early Pregnancy in Goats. Animals (Basel) 2022; 12:2973. [PMID: 36359097 PMCID: PMC9656084 DOI: 10.3390/ani12212973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/07/2023] Open
Abstract
In ruminants, the establishment of pregnancy requires a series of structural and functional changes in the endometrium under the action of hormones, thereby providing an optimal environment for the implantation of the embryo. In this study, we explored the molecular mechanism by which YPEL3 regulates endometrial function during gestation in goats. We found YPEL3 expression was significantly downregulated during early gestation and that YPEL3 overexpression inhibited the expression of ISG15, but had no significant effects on the expression of RSAD2 and CXCL10 in goat endometrial epithelial cells (gEECs). In addition, YPEL3 silencing significantly inhibited PGF2α secretion and the expression of the prostaglandin synthesis-related rate-limiting enzyme-encoding genes PGFS and PTGES, with no significant effect on the expression of PTGS1 and PTGS2. Moreover, YPEL3 inhibited the expression of vimentin and β-catenin and pretreatment of gEECs with the β-catenin activator CHIR99021 prevented a YPEL3-induced decrease in vimentin expression. Collectively, our findings confirm that, as a hormone-regulated factor, YPEL3 regulates endometrial function by inhibiting the Wnt/β-catenin signaling pathway and provide new insights for further clarification of the mechanism by which YPEL3 functions during early pregnancy in ruminants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yaping Jin
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
31
|
Trelford CB, Dagnino L, Di Guglielmo GM. Transforming growth factor-β in tumour development. Front Mol Biosci 2022; 9:991612. [PMID: 36267157 PMCID: PMC9577372 DOI: 10.3389/fmolb.2022.991612] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/15/2022] [Indexed: 11/14/2022] Open
Abstract
Transforming growth factor-β (TGFβ) is a ubiquitous cytokine essential for embryonic development and postnatal tissue homeostasis. TGFβ signalling regulates several biological processes including cell growth, proliferation, apoptosis, immune function, and tissue repair following injury. Aberrant TGFβ signalling has been implicated in tumour progression and metastasis. Tumour cells, in conjunction with their microenvironment, may augment tumourigenesis using TGFβ to induce epithelial-mesenchymal transition, angiogenesis, lymphangiogenesis, immune suppression, and autophagy. Therapies that target TGFβ synthesis, TGFβ-TGFβ receptor complexes or TGFβ receptor kinase activity have proven successful in tissue culture and in animal models, yet, due to limited understanding of TGFβ biology, the outcomes of clinical trials are poor. Here, we review TGFβ signalling pathways, the biology of TGFβ during tumourigenesis, and how protein quality control pathways contribute to the tumour-promoting outcomes of TGFβ signalling.
Collapse
Affiliation(s)
- Charles B. Trelford
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Children’s Health Research Institute and Lawson Health Research Institute, London, ON, Canada
| | - Gianni M. Di Guglielmo
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
32
|
Blanchard N, Link PA, Farkas D, Harmon B, Hudson J, Bogamuwa S, Piper B, Authelet K, Cool CD, Heise RL, Freishtat R, Farkas L. Dichotomous role of integrin-β5 in lung endothelial cells. Pulm Circ 2022; 12:e12156. [PMID: 36438452 PMCID: PMC9684688 DOI: 10.1002/pul2.12156] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/17/2022] [Accepted: 11/07/2022] [Indexed: 11/10/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, devastating disease, and its main histological manifestation is an occlusive pulmonary arteriopathy. One important functional component of PAH is aberrant endothelial cell (EC) function including apoptosis-resistance, unchecked proliferation, and impaired migration. The mechanisms leading to and maintaining physiologic and aberrant EC function are not fully understood. Here, we tested the hypothesis that in PAH, ECs have increased expression of the transmembrane protein integrin-β5, which contributes to migration and survival under physiologic and pathological conditions, but also to endothelial-to-mesenchymal transition (EnMT). We found that elevated integrin-β5 expression in pulmonary artery lesions and lung tissue from PAH patients and rats with PH induced by chronic hypoxia and injection of CD117+ rat lung EC clones. These EC clones exhibited elevated expression of integrin-β5 and its heterodimerization partner integrin-αν and showed accelerated barrier formation. Inhibition of integrin-ανβ5 in vitro partially blocked transforming growth factor (TGF)-β1-induced EnMT gene expression in rat lung control ECs and less in rat lung EC clones and human lung microvascular ECs. Inhibition of integrin-ανβ5 promoted endothelial dysfunction as shown by reduced migration in a scratch assay and increased apoptosis in synergism with TGF-β1. In vivo, blocking of integrin-ανβ5 exaggerated PH induced by chronic hypoxia and CD117+ EC clones in rats. In summary, we found a role for integrin-ανβ5 in lung endothelial survival and migration, but also a partial contribution to TGF-β1-induced EnMT gene expression. Our results suggest that integrin-ανβ5 is required for physiologic function of ECs and lung vascular homeostasis.
Collapse
Affiliation(s)
- Neil Blanchard
- Department of Orthopedic SurgeryUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Patrick A. Link
- Departments of Physiology and Biomedical EngineeringMayo ClinicRochesterMichiganUSA
- Department of Biomedical Engineering, School of EngineeringVirginia Commonwealth UniversityCharlottesvilleVirginiaUSA
| | - Daniela Farkas
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Brennan Harmon
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Jaylen Hudson
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Srimathi Bogamuwa
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Bryce Piper
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Kayla Authelet
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Carlyne D. Cool
- Department of PathologyUniversity of Colorado at DenverDenverColoradoUSA
| | - Rebecca L. Heise
- Department of Biomedical Engineering, School of EngineeringVirginia Commonwealth UniversityCharlottesvilleVirginiaUSA
| | - Robert Freishtat
- Department of Pediatrics, Division of Emergency MedicineChildren's National Health SystemWashingtonDistrict of ColumbiaUSA
| | - Laszlo Farkas
- Division of Pulmonary Disease, College of Medicine, Department of Internal Medicine, Critical Care & Sleep Medicine, Davis Heart and Lung Research InstituteThe Ohio State University Wexner Medical CenterColumbusOhioUSA
- Department of Physiology and BiophysicsVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
33
|
DUSP4 Silencing Enhances the Sensitivity of Breast Cancer Cells to Doxorubicin through the Activation of the JNK/c-Jun Signalling Pathway. Molecules 2022; 27:molecules27196146. [PMID: 36234680 PMCID: PMC9572343 DOI: 10.3390/molecules27196146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/17/2022] [Accepted: 09/02/2022] [Indexed: 11/26/2022] Open
Abstract
Doxorubicin (Dox) has limited efficiency in breast cancer (BC) due to drug-acquired resistance. The epithelial–mesenchymal transition (EMT) plays a major role in the survival and drug resistance of cancer cells. It was suggested that the JNK pathway was implicated in the response to Dox by regulating EMT. DUSP4/or MKP-2 is a well-known regulator of the JNK pathway and was found to be highly expressed in BC. However, its functional significance is not yet fully understood. In the present study, the possible involvement of MKP-2 in Dox-induced EMT was investigated in breast cancer cells. Immunohistochemistry for tissues obtained from BC patients (n = 108) revealed 71.3% of tissues stained positively for MKP-2 while only 28.7% stained negatively. However, MKP-2 protein expression exhibited no significant relationship between BC prognostic factors, such as histological grade, histological type, hormonal status, and Ki-67 marker, its expression was significantly correlated with age 40 or below. In MDA-MB-231 cells, Dox-induced phosphorylation of JNK was sufficiently enhanced in MKP-2 silenced cells. This resulted in the attenuation of Dox-induced EMT, cell cycle arrest, and ultimately accelerated apoptosis. It was confirmed that the acquisition of Dox sensitivity by MKP-2 silencing largely depends on the stimulation of the JNK pathway. Indeed, results showed that overexpressing MKP-2 in non-tumorigenic MCF-12A cells dramatically inhibited Dox-induced JNK activation and, subsequently, cell death. The present study, to our knowledge, is the first to provide evidence for the potential role of MKP-2 in chemoresistance to Dox through modulating the JNK pathway and enhancing EMT.
Collapse
|
34
|
Chen F, Yue LL, Ntsobe TE, Qin LL, Zeng Y, Xie MF, Huang HJ, Peng W, Zeng LS, Liu HJ, Liu Q. Endothelial mesenchymal transformation and relationship with vascular abnormalities. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2022. [DOI: 10.1016/j.jrras.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
35
|
Chen X, Xia X, Dong T, Lin Z, Du L, Zhou H. Trimetazidine Reduces Cardiac Fibrosis in Rats by Inhibiting NOX2-Mediated Endothelial-to-Mesenchymal Transition. Drug Des Devel Ther 2022; 16:2517-2527. [PMID: 35946039 PMCID: PMC9357386 DOI: 10.2147/dddt.s360283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 07/23/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Methods Results Conclusion
Collapse
Affiliation(s)
- Xingxing Chen
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Xue Xia
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Tiancheng Dong
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Zhiwei Lin
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Leilei Du
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Hao Zhou
- Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Correspondence: Hao Zhou, Department of Cardiology, the First Affiliated Hospital of Wenzhou Medical University, NanBai Xiang Avenue, Ouhai District, Wenzhou, 325000, People’s Republic of China, Tel +86 1 396 880 1939, Fax +86 577 555 79796, Email
| |
Collapse
|
36
|
Huang M, Yang F, Zhang D, Lin M, Duan H, El-Mayta R, Zhang L, Qin L, Shewale SV, Pei L, Mitchell MJ, Rader DJ, Fan Y, Gong Y. Endothelial plasticity drives aberrant vascularization and impedes cardiac repair after myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2022; 1:372-388. [PMID: 35571674 PMCID: PMC9104847 DOI: 10.1038/s44161-022-00047-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 03/07/2022] [Indexed: 01/09/2023]
Abstract
Myocardial infarction (MI) is a leading cause of death worldwide, largely because efficient interventions to restore cardiac function after MI are currently lacking. Here, we characterize vascular aberrancies induced by MI, and propose to target acquired endothelial cell (EC) changes to normalize vessels and promote cardiac repair after MI. Single-cell transcriptome analyses of MI-associated ECs indicates that ECs acquire mesenchymal gene signature that result in phenotypic and functional changes and lead to vessel abnormalities. We identify a PDGF/NF-κB/HIF-1α axis that induces Snail expression and mesenchymal phenotypes in ECs under hypoxia, altogether causing aberrant vascularization. EC-specific knockout of PDGFR-β, pharmacological PDGFR inhibition or nanoparticle-based targeted PDGFR-β siRNA delivery in mice attenuates vascular abnormalities in the infarcted tissue and improves cardiac repair after MI. These findings illustrate a mechanism controlling aberrant neovascularization after ischemia, and suggest that targeting PDGF/Snail-mediated endothelial plasticity may offer opportunities for normalizing vasculature and treating ischemic heart diseases.
Collapse
Affiliation(s)
- Menggui Huang
- Department of Radiation Oncology, University of -Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Fan Yang
- Department of Radiation Oncology, University of -Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Duo Zhang
- Department of Radiation Oncology, University of -Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Maohuan Lin
- Division of Human Genetics and Translational Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Hao Duan
- Department of Radiation Oncology, University of -Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, Pennsylvania, USA 19104
| | - Lin Zhang
- Department of Obstetrics & Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Swapnil V. Shewale
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Liming Pei
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Center for Mitochondrial and Epigenomic Medicine, Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA 19104
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, Pennsylvania, USA 19104
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Institute for Regenerative Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Daniel J. Rader
- Division of Human Genetics and Translational Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Yi Fan
- Department of Radiation Oncology, University of -Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| | - Yanqing Gong
- Division of Human Genetics and Translational Medicine, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
- Cardiovascular Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA 19104
| |
Collapse
|
37
|
Chen X, Sun L, Li D, Lai X, Wen S, Chen R, Zhang Z, Li Q, Sun S. Green tea peptides ameliorate diabetic nephropathy by inhibiting the TGF-β/Smad signaling pathway in mice. Food Funct 2022; 13:3258-3270. [PMID: 35234233 DOI: 10.1039/d1fo03615g] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Diabetic nephropathy (DN) is the most important cause of middle and late-stage chronic kidney disease. Green tea polypeptides are extracted from tea pomace, and exhibit various pharmacological effects. In this study, we analyzed the reno-protective effects of green tea peptides in diabetic db/db mice, and explored the underlying mechanisms. Peptide treatment for 5 weeks significantly reduced the blood glucose levels and other indices of diabetes, and alleviated renal injury measured in terms of blood creatinine, urea nitrogen and urinary albumin/urinary creatinine levels. Mechanistically, the green tea peptides downregulated p-Smad2/3, α-SMA, ZO-1 and vimentin proteins in the kidney tissues, and elevated Smad7. Thus, green tea peptides inhibited the deposition of ECM proteins by suppressing excessive activation of the TGF-β/Smad signaling pathway and reducing fibronectin levels. On the other hand, tea peptides ameliorated renal injury by inhibiting the production of inflammatory factors (iNOS and TNF-α) by suppressing the NF-κB signaling pathway. In addition, we confirmed the inhibitory effect of green tea peptides on the TGF-β/Smad signaling pathway in TGF-β1-stimulated HK-2 cells. Therefore, tea peptides can be considered as an effective candidate for alleviating DN.
Collapse
Affiliation(s)
- Xuhui Chen
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China.
| | - Lingli Sun
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| | - Dongli Li
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China. .,International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, China
| | - Xingfei Lai
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| | - Shuai Wen
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| | - Ruohong Chen
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| | - Zhenbiao Zhang
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| | - Qiuhua Li
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| | - Shili Sun
- Tea Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Tea Plant Resources Innovation & Utilization, Guangzhou 510640, China.
| |
Collapse
|
38
|
Ikushima E, Ishikane S, Kishigami T, Matsunaga H, Igawa K, Tomooka K, Nishimura Y, Takahashi-Yanaga F. 2,5-Dimethylcelecoxib attenuates cardiac fibrosis caused by cryoinjury-induced myocardial infarction by suppressing the fibroblast-to-myofibroblast transformation via inhibition of the TGF-β signaling pathway. Biochem Pharmacol 2022; 197:114950. [PMID: 35143754 DOI: 10.1016/j.bcp.2022.114950] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/25/2022]
Abstract
We previously reported that 2,5-dimethylcelecoxib (DM-C), a derivative of celecoxib, lacks cyclooxygenase-2 inhibitory effects and suppresses cardiac remodeling by activating glycogen synthase kinase-3 (GSK-3). However, it remains unclear whether DM-C attenuates fibroblast-to-myofibroblast transformation (FMT), which plays a key role in cardiac fibrosis. Therefore, we evaluated the effect of DM-C on FMT using a cryoinjury-induced myocardial infarction (CMI) mouse model. We found that DM-C attenuated the deterioration of left ventricular ejection fraction after CMI by decreasing cardiac fibrosis. Analysis of the expression level of α-smooth muscle actin (α-SMA), a marker for myofibroblasts, indicated that DM-C decreased FMT at the cardiac injury site. To investigate the mechanism by which DM-C attenuated FMT, fibroblasts obtained from the heart were stimulated with TGF-β to induce FMT, and the effect of DM-C was analyzed. DM-C suppressed the expression of α-SMA and the phosphorylation levels of Smad 2/3 and GSK-3, indicating that DM-C suppressed α-SMA expression by inhibiting the transforming growth factor (TGF)-β signaling pathway via activation of GSK-3. DM-C decreased the expression of collagen, connective tissue growth factor (CTGF) and Snail, which are also known to accelerate cardiac fibrosis. These results suggested that DM-C attenuated cardiac fibrosis by suppressing FMT at the injured site after CMI by inhibiting the TGF-β signaling pathway via activation of GSK-3. Thus, DM-C has potential against cardiac disease as a novel anti-fibrotic agent.
Collapse
Affiliation(s)
- Eigo Ikushima
- Department of Pharmacology, Graduate School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan; Department of Cardiovascular Surgery, School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Shin Ishikane
- Department of Pharmacology, Graduate School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Takehiro Kishigami
- Department of Pharmacology, Graduate School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan; Department of Cardiovascular Surgery, School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Hiroaki Matsunaga
- Department of Pharmacology, Graduate School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Kazunobu Igawa
- Department of Molecular and Material Science, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| | - Katsuhiko Tomooka
- Department of Molecular and Material Science, Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| | - Yosuke Nishimura
- Department of Cardiovascular Surgery, School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan
| | - Fumi Takahashi-Yanaga
- Department of Pharmacology, Graduate School of Medicine, University of Occupational and Environmental Health, Japan, Fukuoka, Japan.
| |
Collapse
|
39
|
Yoshimatsu Y, Watabe T. Emerging roles of inflammation-mediated endothelial–mesenchymal transition in health and disease. Inflamm Regen 2022; 42:9. [PMID: 35130955 PMCID: PMC8818500 DOI: 10.1186/s41232-021-00186-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022] Open
Abstract
Endothelial–mesenchymal transition (EndoMT), a cellular differentiation process in which endothelial cells (ECs) lose their properties and differentiate into mesenchymal cells, has been observed not only during development but also in various pathological states in adults, including cancer progression and organ/tissue fibrosis. Transforming growth factor-β (TGF-β), an inflammation-related cytokine, has been shown to play central roles in the induction of EndoMT. TGF-β induces EndoMT by regulating the expression of various transcription factors, signaling molecules, and cellular components that confer ECs with mesenchymal characteristics. However, TGF-β by itself is not necessarily sufficient to induce EndoMT to promote the progression of EndoMT-related diseases to a refractory extent. In addition to TGF-β, additional activation by other inflammatory factors is often required to stabilize the progression of EndoMT. Since recent lines of evidence indicate that inflammatory signaling molecules act as enhancers of EndoMT, we summarize the roles of inflammatory factors in the induction of EndoMT and related diseases. We hope that this review will help to develop therapeutic strategies for EndoMT-related diseases by targeting inflammation-mediated EndoMT.
Collapse
|
40
|
Oghbaei F, Zarezadeh R, Jafari-Gharabaghlou D, Ranjbar M, Nouri M, Fattahi A, Imakawa K. Epithelial-mesenchymal transition process during embryo implantation. Cell Tissue Res 2022; 388:1-17. [PMID: 35024964 DOI: 10.1007/s00441-021-03574-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/29/2021] [Indexed: 03/01/2023]
Abstract
The epithelial to mesenchymal transition (EMT) in endometrial epithelial and trophectoderm cells is essential for the progression of embryo implantation and its impairment could cause implantation failure. Therefore, EMT should be tightly regulated in both embryonic and endometrial cells during implantation. Studies reported the involvement of numerous factors in EMT regulation, including hormones, growth factors, transcription factors, microRNAs, aquaporins (AQPs), and ion channels. These factors act through different signaling pathways to affect the expression of epithelial and mesenchymal markers as well as the cellular cytoskeleton. Although the mechanisms involved in cancer cell EMT have been well studied, little is known about EMT during embryo implantation. Therefore, we comprehensively reviewed different factors that regulate the EMT, a key event required for the conceptus implantation to the endometrium.Summary sentence: Abnormal epithelial-mesenchymal transition (EMT) process within endometrial epithelial cells (EECs) or trophoblast cells can cause implantation failure. This process is regulated by various factors. Thus, the objective of this review was to summarize the effective factors on the EMT process during implantation.
Collapse
Affiliation(s)
- Farnaz Oghbaei
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Minoo Ranjbar
- Department of Midwifery, Bonab Branch, Islamic Azad University, Bonab, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kazuhiko Imakawa
- Laboratory of Molecular Reproduction, Research Institute of Agriculture, Tokai University, Kumamoto, 862-8652, Japan
| |
Collapse
|
41
|
Pei B, Zhang N, Pang T, Sun G. Linagliptin ameliorates pulmonary fibrosis in systemic sclerosis mouse model via inhibition of endothelial-to-mesenchymal transition. Mol Cell Biochem 2022; 477:995-1007. [PMID: 34988855 DOI: 10.1007/s11010-021-04349-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/22/2021] [Indexed: 12/27/2022]
Abstract
Systemic sclerosis (SSc) is a connective tissue disease that often causes pulmonary fibrosis. Dipeptidyl peptidase 4 (DPP4) inhibitor has shown anti-fibrotic properties in various fibrotic diseases. However, only two studies have reported its anti-fibrosis effects in pulmonary fibrosis, and the mechanism is not completely clear. In the present study, we further investigated the protective effects of linagliptin, a highly specific DPP4 inhibitor, on pulmonary fibrosis in SSc mouse model and the potential mechanisms. The results showed that linagliptin ameliorated pulmonary fibrosis in SSc mouse model, as evidenced by improved pathological changes of lung and body weight loss induced by BLM. Linagliptin also reduced BLM-induced oxidative stress, inflammation in lung in vivo. We revealed that linagliptin attenuated BLM-induced endothelial-to-mesenchymal transition (EndMT) in vitro and in vivo. BLM-induced enhanced migration ability of endothelial cells was also alleviated by linagliptin. Moreover, we confirmed that the Akt/mammalian target of rapamycin pathway was involved in BLM-induced EndMT in vivo, which was suppressed by linagliptin. In summary, we further confirmed the therapeutic effects of linagliptin on pulmonary fibrosis in SSc mouse model, which is based on its inhibitory effects on EndMT, oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Biwei Pei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Na Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Tingting Pang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
42
|
CXCL4 drives fibrosis by promoting several key cellular and molecular processes. Cell Rep 2022; 38:110189. [PMID: 34986347 DOI: 10.1016/j.celrep.2021.110189] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 02/28/2021] [Accepted: 12/08/2021] [Indexed: 01/05/2023] Open
Abstract
Fibrosis is a major cause of mortality worldwide, characterized by myofibroblast activation and excessive extracellular matrix deposition. Systemic sclerosis is a prototypic fibrotic disease in which CXCL4 is increased and strongly correlates with skin and lung fibrosis. Here we aim to elucidate the role of CXCL4 in fibrosis development. CXCL4 levels are increased in multiple inflammatory and fibrotic mouse models, and, using CXCL4-deficient mice, we demonstrate the essential role of CXCL4 in promoting fibrotic events in the skin, lungs, and heart. Overexpressing human CXCL4 in mice aggravates, whereas blocking CXCL4 reduces, bleomycin-induced fibrosis. Single-cell ligand-receptor analysis predicts CXCL4 to affect endothelial cells and fibroblasts. In vitro, we confirm that CXCL4 directly induces myofibroblast differentiation and collagen synthesis in different precursor cells, including endothelial cells, by stimulating endothelial-to-mesenchymal transition. Our findings identify a pivotal role of CXCL4 in fibrosis, further substantiating the potential role of neutralizing CXCL4 as a therapeutic strategy.
Collapse
|
43
|
Rogers MP, Mi Z, Li NY, Wai PY, Kuo PC. Tumor: Stroma Interaction and Cancer. EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:59-87. [PMID: 35165860 DOI: 10.1007/978-3-030-91311-3_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The understanding of how normal cells transform into tumor cells and progress to invasive cancer and metastases continues to evolve. The tumor mass is comprised of a heterogeneous population of cells that include recruited host immune cells, stromal cells, matrix components, and endothelial cells. This tumor microenvironment plays a fundamental role in the acquisition of hallmark traits, and has been the intense focus of current research. A key regulatory mechanism triggered by these tumor-stroma interactions includes processes that resemble epithelial-mesenchymal transition, a physiologic program that allows a polarized epithelial cell to undergo biochemical and cellular changes and adopt mesenchymal cell characteristics. These cellular adaptations facilitate enhanced migratory capacity, invasiveness, elevated resistance to apoptosis, and greatly increased production of ECM components. Indeed, it has been postulated that cancer cells undergo epithelial-mesenchymal transition to invade and metastasize.In the following discussion, the physiology of chronic inflammation, wound healing, fibrosis, and tumor invasion will be explored. The key regulatory cytokines, transforming growth factor β and osteopontin, and their roles in cancer metastasis will be highlighted.
Collapse
Affiliation(s)
- Michael P Rogers
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Zhiyong Mi
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Neill Y Li
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Philip Y Wai
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Paul C Kuo
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
44
|
Li XL, Liu J, Chen XS, Cheng LM, Liu WL, Chen XF, Li YJ, Guan YY, Zeng X, Du YH. Blockade of TMEM16A protects against renal fibrosis by reducing intracellular Cl - concentration. Br J Pharmacol 2021; 179:3043-3060. [PMID: 34961937 DOI: 10.1111/bph.15786] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 09/27/2021] [Accepted: 12/16/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Renal fibrosis is the final common outcome in most forms of CKD. However, the underlying causal mechanisms remain obscure. The present study examined whether TMEM16A, a Ca2+ -activated chloride channel, contributes to the progress of renal fibrosis. EXPERIMENTAL APPROACH Masson staining, western blot and immunohistochemistry were used to measure renal fibrosis and related proteins expression. MQAE was used to evaluate the intracellular Cl- concentration. KEY RESULTS TMEM16A expression was significantly upregulated in fibrotic kidneys of unilateral ureteral obstruction (UUO) and high-fat diet murine models, and in renal samples of IgA nephropathy patients. In vivo knockdown of TMEM16A with adenovirus harboring TMEM16A-shRNA or inhibition of TMEM16A channel activity with its specific inhibitor CaCCinh-A01 or T16Ainh-A01 effectively prevented UUO-induced renal fibrosis and decreased protein expression of fibronectin, α-SMA and collagen in the obstructed kidneys. In cultured HK2 cells, knockdown or inhibition of TMEM16A suppressed TGF-β1-induced epithelial to mesenchymal transition, reduced snail1 expression and phosphorylation of Smad2/3 and ERK1/2, whereas overexpression of TMEM16A showed the opposite effects. TGF-β1 increased [Cl- ]i in HK2 cells, which was inhibited by knockdown or inhibition of TMEM16A. Reducing [Cl- ]i by low Cl- culture medium significantly blunted TGF-β1-induced Smad2/3 phosphorylation and profibrotic factors expression. The profibrotic effects of TGF-β1 were also abrogated by the inhibitor of SGK1, a kinase whose activity was also suppressed by reducing [Cl- ]i. CONCLUSION AND IMPLICATIONS Blockade of TMEM16A prevented the progression of kidney fibrosis, likely by suppressing [Cl- ]i/SGK1/TGF-β1 signaling pathway. TMEM16A may be a potential new therapeutic target against renal fibrosis.
Collapse
Affiliation(s)
- Xiao-Long Li
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jing Liu
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiao-Shan Chen
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Li-Min Cheng
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wei-Ling Liu
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xing-Feng Chen
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yue-Jiao Li
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yong-Yuan Guan
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xin Zeng
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yan-Hua Du
- Department of Pharmacology, Cardiac & Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Chen J, Han S, Chen J, Hu P, Zeng Z, Hu Y, Xiong H, Ke Z, Zhang Y, Xu F, Zhao G. A reciprocal feedback of miR-548ac/YB-1/Snail induces EndMT of HUVECs during acidity microenvironment. Cancer Cell Int 2021; 21:692. [PMID: 34930270 PMCID: PMC8691019 DOI: 10.1186/s12935-021-02388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/02/2021] [Indexed: 11/10/2022] Open
Abstract
Background Researches indicated the process of Endothelial-Mesenchymal-Transition (EndMT) of vascular endothelial cells (ECs) was critically involved in the progression of tumor. ECs demonstrated functional and phenotypic heterogeneity when located under different microenvironments. The extracellular pH of tumor tissues was acidic compared to that of normal tissues. However, there was still unclear whether the acidic microenvironment affected the EndMT of vascular ECs. Methods Human Umbilical Vein Endothelial Cell (HUVECs) was cultured under the normal or acidic medium to evaluate the alteration of morphology, migration, permeability, and EndMT markers. Microarray assay was adopted to analyze the differential expression of miRNAs in the acidity-treated HUVECs. Gain- and loss- of function experiments were performed to evaluate the functional role of miRNA-548ac on acidity-induced EndMT of HUVECs. Luciferase reporter and Chromatin-immunoprecipitation assays were conducted to assess the downstream pathway of miRNA-548ac in acidity-induced EndMT of HUVECs. Results Our results showed that HUVECs demonstrated mesenchymal transition under acidic conditions with the increase of migration, permeability, and expression of α-SMA and Vimentin, but the expression of vascular endothelial cadherin (VE-cadherin) and CD31 were reduced. In addition, the acidity-treated HUVECs remarkably facilitated the transmigration of pancreatic cancer cells. The expression of miRNA-548ac was significantly decreased in the acidity-treated HUVECs. Moreover, overexpression of miR-548ac inhibited the EndMT of HUVECs and consequently impeded the transmigration of pancreatic cancer cells. The miR-548ac inhibited the expression of YB-1 by binding to the 3’UTR of its mRNA, and YB-1 promoted the translation of Snail which was a critical regulator of EndMT. What’s more, Snail transcriptionally inhibited the expression of miR-548ac through binding to the promoter of its host gene. Conclusions Our data implicated that the acidic microenvironment promoted the EndMT of HUVECs by the miR-548ac/YB-1/Snail axis, which could contribute to the metastasis of pancreatic cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02388-8.
Collapse
Affiliation(s)
- Jingyuan Chen
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shengbo Han
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinhuang Chen
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Hu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhu Zeng
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuhang Hu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hewei Xiong
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zunxiang Ke
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ya Zhang
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fengyu Xu
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Gang Zhao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
46
|
Kochetkova M, Samuel MS. Differentiation of the tumor microenvironment: are CAFs the Organizer? Trends Cell Biol 2021; 32:285-294. [PMID: 34895986 DOI: 10.1016/j.tcb.2021.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Cancers contain a suite of genetically stable cells within an extracellular matrix, collectively termed the tumor microenvironment (TME). The TME strongly influences disease outcome for patients. Gleaning clues from the literature, we propose that the TME should be viewed not as disparate populations of cells constituting a pathological lesion, but as a cohesive tissue constituting a novel pathological organ, arising from the coordinated differentiation of its constituent cell types - a process we have termed tumor-associated neodifferentiation (TAND). We also discuss why cancer-associated fibroblasts (CAFs) may assume the role of Organizer of this organ, directing the recruitment and differentiation of cells within the TME. Viewing the microenvironment in this way will reveal new cancer vulnerabilities that may be exploited for therapy.
Collapse
Affiliation(s)
- Marina Kochetkova
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
47
|
Zhang Y, Arner EN, Rizvi A, Toombs JE, Huang H, Warner SL, Foulks JM, Brekken RA. AXL inhibitor TP-0903 reduces metastasis and therapy resistance in pancreatic cancer. Mol Cancer Ther 2021; 21:38-47. [PMID: 34675118 DOI: 10.1158/1535-7163.mct-21-0293] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 08/19/2021] [Accepted: 10/19/2021] [Indexed: 11/16/2022]
Abstract
Pancreatic cancer is the 3rd leading cause of cancer-related deaths in the United States with a 5-year survival less than 5%. Resistance to standard therapy and limited response to immune checkpoint blockade due to the immunosuppressive and stroma-rich microenvironment remain major challenges in the treatment of pancreatic cancer. A key cellular program involved in therapy resistance is epithelial plasticity, which is also associated with invasion, metastasis, and evasion of immune surveillance. The receptor tyrosine kinase AXL is a key driver of tumor cell epithelial plasticity. High expression and activity of AXL is associated with poor prognosis, metastasis, and therapy resistance in multiple types of cancer including pancreatic. Here, we show that an AXL inhibitor (TP-0903), has anti-tumor and therapy sensitizing effects in pre-clinical models of pancreatic ductal adenocarcinoma (PDA). We demonstrate that TP-0903 as a single agent or in combination with gemcitabine and/or anti-programmed cell death protein 1 (PD1) antibody has anti-metastatic and anti-tumor effects in PDA tumor bearing mice, leading to increased survival. Additionally, gene expression analysis of tumors demonstrated upregulation of pro-inflammatory and immune activation genes in tumors from TP-0903-treated animals compared to the vehicle, indicating pharmacologic inhibition of AXL activation leads to an immunostimulatory microenvironment. This effect was augmented when TP-0903 was combined with gemcitabine and anti-PD1 antibody. These results provide clear rationale for evaluating TP-0903 in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yuqing Zhang
- Hamon Center for Therapeutic Oncology Research, Dallas, TX
- Cancer Biology Graduate Program, Dallas, TX
- Department of Surgery, Dallas, TX
| | - Emily N Arner
- Hamon Center for Therapeutic Oncology Research, Dallas, TX
- Cancer Biology Graduate Program, Dallas, TX
- Department of Surgery, Dallas, TX
| | - Ali Rizvi
- Hamon Center for Therapeutic Oncology Research, Dallas, TX
| | - Jason E Toombs
- Hamon Center for Therapeutic Oncology Research, Dallas, TX
- Department of Surgery, Dallas, TX
| | - Huocong Huang
- Hamon Center for Therapeutic Oncology Research, Dallas, TX
- Department of Surgery, Dallas, TX
| | | | | | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, Dallas, TX.
- Cancer Biology Graduate Program, Dallas, TX
- Department of Surgery, Dallas, TX
- Department of Pharmacology, UT Southwestern, Dallas, Texas
| |
Collapse
|
48
|
Fang JS, Hultgren NW, Hughes CCW. Regulation of Partial and Reversible Endothelial-to-Mesenchymal Transition in Angiogenesis. Front Cell Dev Biol 2021; 9:702021. [PMID: 34692672 PMCID: PMC8529039 DOI: 10.3389/fcell.2021.702021] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022] Open
Abstract
During development and in several diseases, endothelial cells (EC) can undergo complete endothelial-to-mesenchymal transition (EndoMT or EndMT) to generate endothelial-derived mesenchymal cells. Emerging evidence suggests that ECs can also undergo a partial EndoMT to generate cells with intermediate endothelial- and mesenchymal-character. This partial EndoMT event is transient, reversible, and supports both developmental and pathological angiogenesis. Here, we discuss possible regulatory mechanisms that may control the EndoMT program to dictate whether cells undergo complete or partial mesenchymal transition, and we further consider how these pathways might be targeted therapeutically in cancer.
Collapse
Affiliation(s)
- Jennifer S. Fang
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| | - Nan W. Hultgren
- Department of Ophthalmology, Stein Eye Institute, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Christopher C. W. Hughes
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
49
|
Endothelial Heterogeneity in Development and Wound Healing. Cells 2021; 10:cells10092338. [PMID: 34571987 PMCID: PMC8469713 DOI: 10.3390/cells10092338] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/30/2021] [Accepted: 09/06/2021] [Indexed: 12/28/2022] Open
Abstract
The vasculature is comprised of endothelial cells that are heterogeneous in nature. From tissue resident progenitors to mature differentiated endothelial cells, the diversity of these populations allows for the formation, maintenance, and regeneration of the vascular system in development and disease, particularly during situations of wound healing. Additionally, the de-differentiation and plasticity of different endothelial cells, especially their capacity to undergo endothelial to mesenchymal transition, has also garnered significant interest due to its implication in disease progression, with emphasis on scarring and fibrosis. In this review, we will pinpoint the seminal discoveries defining the phenotype and mechanisms of endothelial heterogeneity in development and disease, with a specific focus only on wound healing.
Collapse
|
50
|
Epithelial plasticity, epithelial-mesenchymal transition, and the TGF-β family. Dev Cell 2021; 56:726-746. [PMID: 33756119 DOI: 10.1016/j.devcel.2021.02.028] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/04/2021] [Accepted: 02/23/2021] [Indexed: 12/15/2022]
Abstract
Epithelial cells repress epithelial characteristics and elaborate mesenchymal characteristics to migrate to other locations and acquire new properties. Epithelial plasticity responses are directed through cooperation of signaling pathways, with TGF-β and TGF-β-related proteins playing prominent instructive roles. Epithelial-mesenchymal transitions (EMTs) directed by activin-like molecules, bone morphogenetic proteins, or TGF-β regulate metazoan development and wound healing and drive fibrosis and cancer progression. In carcinomas, diverse EMTs enable stem cell generation, anti-cancer drug resistance, genomic instability, and localized immunosuppression. This review discusses roles of TGF-β and TGF-β-related proteins, and underlying molecular mechanisms, in epithelial plasticity in development and wound healing, fibrosis, and cancer.
Collapse
|