1
|
Liu Q, Yu M, Lin Z, Wu L, Xia P, Zhu M, Huang B, Wu W, Zhang R, Li K, Zhu L, Wang Q. COL1A1-positive endothelial cells promote gastric cancer progression via the ANGPTL4-SDC4 axis driven by endothelial-to-mesenchymal transition. Cancer Lett 2025; 623:217731. [PMID: 40254092 DOI: 10.1016/j.canlet.2025.217731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/19/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer (GC) is an aggressive and heterogeneous disease with poor survival outcomes. The progression of GC involves complex, multi-step processes. Endothelial cells (ECs) play a crucial role in tumor angiogenesis, proliferation, invasion, and metastasis, particularly through the process of endothelial-to-mesenchymal transition (EndoMT). However, the specific role and mechanisms of EndoMT in gastric cancer remain unclear. Based on 6 GC single-cell RNA-sequencing (scRNA-seq) cohorts (samples = 97), we established an EndoMT-related gene signature, termed EdMTS. Leveraging this gene signature, ssGSEA was applied to calculate sample scores across multiple bulk RNA-seq datasets, which include information on immunotherapy, metastasis, GC progression, and survival. Moreover, we applied the Monocle2 method to calculate cell pseudotime and used CellChat to analyze interactions between malignant and EC cells. We verified the molecular mechanism by multiple immunofluorescence and cell function experiments. Findings In this study, we established a single-cell atlas of ECs in GC and identified a subpopulation of COL1A1+ ECs that play a critical role in tumor progression and metastasis. These COL1A1+ ECs were significantly associated with worse clinical outcomes in GC patients. Further analysis revealed that COL1A1+ ECs originated from lymphatic ECs and underwent EndoMT through the upregulation of CEBPB, driving tumor invasiveness. Moreover, COL1A1+ ECs interacted with malignant cells via ANGPTL4-SDC4 axis, enhancing invasion and migration. These findings provide a deeper understanding of the role of COL1A1+ ECs in GC progression and highlight potential therapeutic targets for disrupting the EndoMT process in these cells to provide a benefit for GC patients.
Collapse
Affiliation(s)
- Quanzhong Liu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Miao Yu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Zihan Lin
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Lingxiang Wu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Peng Xia
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Mengyan Zhu
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Bin Huang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Wei Wu
- School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Ruohan Zhang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Kening Li
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China
| | - Lingjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Qianghu Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China; The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, 210002, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Byford AR, Fakonti G, Shao Z, Soni S, Earle SL, Bajarwan M, Morley LC, Holder B, Scott EM, Forbes K. Endothelial-to-mesenchymal transition in the fetoplacental macrovasculature and microvasculature in pregnancies complicated by gestational diabetes. J Physiol 2025. [PMID: 40349322 DOI: 10.1113/jp287931] [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: 11/14/2024] [Revised: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Gestational diabetes mellitus (GDM) is linked to altered fetal development and an increased risk of offspring developing cardiometabolic diseases in adulthood. The mechanisms responsible are unclear; however, GDM is associated with altered fetoplacental vascularisation, fibrosis and endothelial dysfunction. In non-pregnant individuals with diabetes, similar vascular changes are attributed to disruptions in endothelial-to-mesenchymal transition (EndMT), a key process where endothelial cells adopt a mesenchymal phenotype. Here, we assess whether alterations in the fetoplacental macro- and microvasculature are attributed to EndMT, using human umbilical vein endothelial cells (HUVECs) and human term placental tissue, respectively. Transforming growth factor (TGF)-β2 and interleukin (IL)-1β induced morphological and molecular changes consistent with EndMT in both GDM and non-GDM HUVECs. The ability of TGF-β2 and IL-1β to alter expression of known EndMT regulators, VWF, TGFBR1, IL1B and IL1R1, was diminished in GDM HUVECs; however, all other hallmarks of EndMT were similar. In placental villous tissue, Slug and Snail, two key transcriptional regulators of EndMT, were detected in the villous stroma, suggesting that EndMT probably occurs in the placental microvasculature. We observed a reduction in endothelial marker genes PECAM1, VWF and CDH5 in GDM placentas, suggesting reduced placental vascularisation. This was accompanied by a reduction in EndMT regulators SNAI2, TGB2, TGFB3 and TGFBR2; however, there was no change in mesenchymal markers or other EndMT regulators. This suggests that there may be some alterations in EndMT in GDM but this probably does not fully explain the endothelial dysfunction and altered vascularisation that occurs in the fetoplacental vasculature in pregnancies complicated by GDM. KEY POINTS: Gestational diabetes mellitus (GDM) has been linked to altered placental vascularisation, fibrosis and endothelial dysfunction. Disruptions in endothelial-to-mesenchymal transition (EndMT), a process where endothelial cells adopt a mesenchymal phenotype, has been linked to vascular complications in diabetes, but EndMT in GDM has not been investigated. Transforming growth factor (TGF)-β2 and interleukin (IL)-1β induced morphological and molecular changes consistent with EndMT in GDM and non-GDM human umbilical vein endothelial cells (HUVECs). Although the expression of EndMT mediators, VWF, TGFBR1, IL1B, and IL1R1, was diminished in GDM HUVECs, other EndMT hallmarks were similar. Transcriptional regulators of EndMT, Slug and Snail, were detected in the human term placenta. Despite a reduction in endothelial markers, PECAM1, VWF and CDH5, as well as SNAI2, TGFB2/3 and TGFBR2 in GDM placenta, there was no change in mesenchymal or other EndMT markers. This suggests that, although there may be some changes to EndMT in GDM, the vascular dysfunction is probably not explained fully by alterations in EndMT.
Collapse
Affiliation(s)
- Abigail R Byford
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Georgia Fakonti
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Ziyu Shao
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sharanam Soni
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Sophie L Earle
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Muath Bajarwan
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Lara C Morley
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Beth Holder
- Institute of Reproductive and Developmental Biology (IRDB), Imperial College London, London, UK
| | - Eleanor M Scott
- Clinical and Population Sciences, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Karen Forbes
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| |
Collapse
|
3
|
Jiang H, Zhou Y, Zhang W, Li H, Ma W, Ji X, Zhou C. Molecular mechanisms of endothelial-mesenchymal transition and its pathophysiological feature in cerebrovascular disease. Cell Biosci 2025; 15:49. [PMID: 40253404 PMCID: PMC12008988 DOI: 10.1186/s13578-025-01393-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
The phenomenon of endothelial-mesenchymal transition (EndMT), a distinct subtype of epithelial-mesenchymal transition (EMT), has garnered significant attention from scholars. EndMT refers to the process whereby endothelial cells (ECs) transform into mesenchymal cells in response to various stimuli, resulting in the loss of their original characteristics. This process has diverse implications in both physiological and pathological states. Under physiological conditions, EndMT plays a crucial role in the development of the cardiovascular system. Conversely, under pathological conditions, EndMT has been identified as a pivotal factor in the development of cardiovascular diseases. Nonetheless, a comprehensive overview of EndMT in cerebrovascular disease is currently lacking. Here, we discuss the heterogeneity of EndMT occurrence and the regulatory factors involved in its development and analyze the feasibility of EndMT as a therapeutic target, aiming to provide a solid theoretical foundation and evidence to address diseases caused by pathological EndMT.
Collapse
Affiliation(s)
- Huimin Jiang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China
| | - Yifan Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China
| | - Weiyue Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Hui Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Ma
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China.
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Chen Zhou
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Ministry of Science and Technology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
4
|
Srivastava SP, Kopasz-Gemmen O, Thurman A, Rajendran BK, Selvam MM, Kumar S, Srivastava R, Suresh MX, Kumari R, Goodwin JE, Inoki K. The molecular determinants regulating redox signaling in diabetic endothelial cells. Front Pharmacol 2025; 16:1563047. [PMID: 40290438 PMCID: PMC12023289 DOI: 10.3389/fphar.2025.1563047] [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: 01/18/2025] [Accepted: 03/14/2025] [Indexed: 04/30/2025] Open
Abstract
Oxidation and reduction are vital for keeping life through several prime mechanisms, including respiration, metabolism, and other energy supplies. Mitochondria are considered the cell's powerhouse and use nutrients to produce redox potential and generate ATP and H2O through the process of oxidative phosphorylation by operating electron transfer and proton pumping. Simultaneously, mitochondria also produce oxygen free radicals, called superoxide (O2 -), non-enzymatically, which interacts with other moieties and generate reactive oxygen species (ROS), such as hydrogen peroxide (H2O2), peroxynitrite (ONOO-), and hydroxyl radical (OH-). These reactive oxygen species modify nucleic acids, proteins, and carbohydrates and ultimately cause damage to organs. The nutrient-sensing kinases, such as AMPK and mTOR, function as a key regulator of cellular ROS levels, as loss of AMPK or aberrant activation of mTOR signaling causes ROS production and compromises the cell's oxidant status, resulting in various cellular injuries. The increased ROS not only directly damages DNA, proteins, and lipids but also alters cellular signaling pathways, such as the activation of MAPK or PI3K, the accumulation of HIF-1α in the nucleus, and NFkB-mediated transcription of pro-inflammatory cytokines. These factors cause mesenchymal activation in renal endothelial cells. Here, we discuss the biology of redox signaling that underlies the pathophysiology of diabetic renal endothelial cells.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | | | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
| | - Barani Kumar Rajendran
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, United States
| | - M. Masilamani Selvam
- Department of Pharmaceutical Technology, Paavai Engineering College, Namakkal, Tamil Nadu, India
| | - Sandeep Kumar
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Rohit Srivastava
- Laboratory of Medical Transcriptomics, Department of Endocrinology, Nephrology Services, Hadassah Hebrew-University Medical Center, Jerusalem, Israel
| | - M. Xavier Suresh
- School of Advanced Sciences and Languages, VIT Bhopal University, Sehore, Madhya Pradesh, India
| | - Reena Kumari
- Department of Physiology, Augusta University, Augusta, GA, United States
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutic Program, Yale University School of Medicine, New Haven, CT, United States
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
5
|
Tugizov S. HIV-1 Tat-induced disruption of epithelial junctions and epithelial-mesenchymal transition of oral and genital epithelial cells lead to increased invasiveness of neoplastic cells and the spread of herpes simplex virus and cytomegalovirus. Front Immunol 2025; 16:1541532. [PMID: 40018040 PMCID: PMC11866325 DOI: 10.3389/fimmu.2025.1541532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025] Open
Abstract
Human immunodeficiency virus (HIV-1) transactivator Tat is a unique multi-functional viral protein secreted by infected cells. Although its primary function is to promote HIV-1 transcription, secreted Tat interacts with neighboring cells and induces numerous disease-associated pathological changes. Despite the substantial reduction of viral load and disease burden, Tat expression and secretion persist in people living with HIV who are undergoing treatment with highly effective combination antiretroviral therapy (cART). Tat interacts with both oral and genital epithelial cells and impairs their mucosal barrier functions, which facilitates the entry of other pathogenic viruses. Tat-mediated interactions with both human papillomavirus (HPV) -infected and HPV-negative neoplastic epithelial cells lead to epithelial-mesenchymal transition and increased invasiveness of malignant cells. Likewise, Tat-induced disruption of oral epithelial cell junctions leads to herpes simplex virus-1 (HSV-1) infection and spread via exposure of its receptor, nectin-1. HIV-1 Tat facilitates infection and spread of human cytomegalovirus (HCMV) by activating mitogen-activated protein kinases (MAPK) and promoting NF-κB signaling, both critical for the replication and production of progeny virions. HIV extracellular Tat also plays a critical role in human herpesvirus 8 (HHV8) -caused Kaposi sarcoma (KS) pathogenesis by synergizing with HHV-8 lytic proteins and promoting the proliferation, angiogenesis, and migration of endothelial cells. Collectively, these findings emphasize the critical impact of HIV-1 Tat on HIV/AIDS pathogenesis during the cART era and highlight the need for further research on the molecular mechanisms underlying Tat-mediated interactions with oral and genital mucosal epithelial cells.
Collapse
Affiliation(s)
- Sharof Tugizov
- Department of Medicine, School of Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
6
|
Greenman R, Weston CJ. CCL24 and Fibrosis: A Narrative Review of Existing Evidence and Mechanisms. Cells 2025; 14:105. [PMID: 39851534 PMCID: PMC11763828 DOI: 10.3390/cells14020105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Tissue fibrosis results from a dysregulated and chronic wound healing response accompanied by chronic inflammation and angiogenesis. Regardless of the affected organ, fibrosis shares the following common hallmarks: the recruitment of immune cells, fibroblast activation/proliferation, and excessive extracellular matrix deposition. Chemokines play a pivotal role in initiating and advancing these fibrotic processes. CCL24 (eotaxin-2) is a chemokine secreted by immune cells and epithelial cells, which promotes the trafficking of immune cells and the activation of profibrotic cells through CCR3 receptor binding. Higher levels of CCL24 and CCR3 were found in the tissue and sera of patients with fibro-inflammatory diseases, including primary sclerosing cholangitis (PSC), systemic sclerosis (SSc), and metabolic dysfunction-associated steatohepatitis (MASH). This review delves into the intricate role of CCL24 in fibrotic diseases, highlighting its impact on fibrotic, immune, and vascular pathways. We focus on the preclinical and clinical evidence supporting the therapeutic potential of blocking CCL24 in diseases that involve excessive inflammation and fibrosis.
Collapse
Affiliation(s)
| | - Chris J. Weston
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- National Institute for Health and Care Research (NIHR), Birmingham Biomedical Research Centre, Birmingham B15 2TT, UK
| |
Collapse
|
7
|
Qian C, Dong G, Yang C, Zheng W, Zhong C, Shen Q, Lu Y, Zhao Y. Broadening horizons: molecular mechanisms and disease implications of endothelial-to-mesenchymal transition. Cell Commun Signal 2025; 23:16. [PMID: 39789529 PMCID: PMC11720945 DOI: 10.1186/s12964-025-02028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is defined as an important process of cellular differentiation by which endothelial cells (ECs) are prone to lose their characteristics and transform into mesenchymal cells. During EndMT, reduced expression of endothelial adhesion molecules disrupts intercellular adhesion, triggering cytoskeletal reorganization and mesenchymal transition. Numerous studies have proved that EndMT is a multifaceted biological event driven primarily by cytokines such as TGF-β, TNF-α, and IL-1β, alongside signaling pathways like WNT, Smad, MEK-ERK, and Notch. Nevertheless, the exact roles of EndMT in complicated diseases have not been comprehensively reviewed. In this review, we summarize the predominant molecular regulatory mechanisms and signaling pathways that contribute to the development of EndMT, as well as highlight the contributions of a series of imperative non-coding RNAs in curbing the initiation of EndMT. Furthermore, we discuss the significant impact of EndMT on worsening vasculature-related diseases, including cancer, cardiovascular diseases, atherosclerosis, pulmonary vascular diseases, diabetes-associated fibrotic conditions, and cerebral cavernous malformation, providing the implications that targeting EndMT holds promise as a therapeutic strategy to mitigate disease progression.
Collapse
Affiliation(s)
- Cheng Qian
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanglu Dong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunmei Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weiwei Zheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chongjin Zhong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiuhong Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yang Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
8
|
Abdelsalam SS, Zahid MA, Ghanem SK, Khan A, Parray A, Agouni A. Sestrin2 Suppression Promotes Endothelial-Mesenchymal Transition and Exacerbates Methylglyoxal-Induced Endothelial Dysfunction. Int J Mol Sci 2024; 25:13463. [PMID: 39769227 PMCID: PMC11676724 DOI: 10.3390/ijms252413463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 01/03/2025] Open
Abstract
Sestrin2 (SESN2) is a stress-inducible protein known for its cytoprotective functions, but its role in diabetic vascular complications remains unclear. This study investigated the impact of SESN2 on methylglyoxal (MGO)-induced endothelial-mesenchymal transition (EndMT). Human endothelial cells were transfected with SESN2 siRNA duplexes to silence SESN2 expression, followed by MGO treatment. SESN2 knockdown significantly exacerbated MGO-induced oxidative stress, as evidenced by the reduced expression of antioxidant markers. Furthermore, SESN2 silencing enhanced the inflammatory response to MGO, demonstrated by the increased levels of pro-inflammatory cytokines. Notably, SESN2 deficiency promoted EndMT, a key process in diabetes-induced cardiovascular complications, as shown by the increased expression of mesenchymal markers and the decreased expression of endothelial markers. These findings suggest that SESN2 plays a critical protective role in endothelial cells against MGO-induced damage. The study provides novel insights into the molecular mechanisms underlying diabetic cardiovascular complications and identifies SESN2 as a potential therapeutic target for preventing endothelial dysfunction in diabetes. Our results indicate that SESN2 downregulation may contribute to the pathogenesis of diabetic vascular complications by promoting EndMT, increased oxidative stress, and inflammation.
Collapse
Affiliation(s)
- Shahenda Salah Abdelsalam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (S.S.A.); (M.A.Z.); (S.K.G.)
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (S.S.A.); (M.A.Z.); (S.K.G.)
| | - Sarah Khalaf Ghanem
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (S.S.A.); (M.A.Z.); (S.K.G.)
| | - Abbas Khan
- Department of Biological Sciences, School of Medical and Life Sciences (SMLS), Sunway University, Bandar Sunway 47500, Malaysia;
| | - Aijaz Parray
- The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar;
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar; (S.S.A.); (M.A.Z.); (S.K.G.)
| |
Collapse
|
9
|
Lamont HC, Wright AL, Devries K, Okur KE, Jones M, Masood I, Hill LJ, Nazhat SN, Grover LM, Haj AJE, Metcalfe AD. Trabecular meshwork cell differentiation in response to collagen and TGFβ-2 spatial interactions. Acta Biomater 2024; 189:217-231. [PMID: 39218278 DOI: 10.1016/j.actbio.2024.08.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Primary open-angle glaucoma (POAG) is currently the most prevalent cause of irreversible blindness globally. To date, few in vitro models that can faithfully recapitulate the complex architecture of the trabecular meshwork (TM) and the specialised trabecular meshwork cell (TMC) characteristics that are local to the structurally opposing regions. This study aimed to investigate the parameters that govern TMC phenotype by adapting the extracellular matrix structure to mimic the juxtacanalicular tissue (JCT) region of the TM. Initially, TMC phenotypic characteristics were investigated within type I collagen matrices of controlled fiber density and anisotropy, generated through confined plastic compression (PC). Notably, PC-collagen presented biophysical cues that induced JCT cellular characteristics (elastin, α-β-Crystallin protein expression, cytoskeletal remodelling, increased mesenchymal markers and JCT-specific genetic markers). In parallel, a pathological mesenchymal phenotype associated with POAG was induced through localised transforming growth factor -beta 2 (TGFβ-2) exposure. This resulted in a profile of alternative mesenchymal states (fibroblast/smooth muscle or myofibroblast) displayed by the TMC in vitro. Overall, the study provides an advanced insight into the biophysical cues that modulate TMC fate, inducing a JCT-specific phenotype and transient mesenchymal characteristics that reflect healthy and pathological scenarios. STATEMENT OF SIGNIFICANCE: Glaucoma is a leading cause of blindness, with a lack of long-term efficacy within current drug candidates. Reliable trabecular meshwork (TM) in vitro models will be critical for enhancing the fields understanding of healthy and disease states for pre-clinical testing. Trabecular meshwork cells (TMCs) display heterogeneity throughout the hierarchical TM, however our understanding into recapitulating these phenotypes in vitro, remains elusive. This study hypothesizes the importance of specific matrix/growth factor spatial stimuli in governing TMCs phenotype. By emulating certain biophysical/biochemical in vivo parameters, we introduce an advanced profile of distinct TMC phenotypic states, reflecting healthy and disease scenarios. A notion that has not be stated prior and a fundamental consideration for future 3D TM in vitro modelling.
Collapse
Affiliation(s)
- Hannah C Lamont
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, UK.
| | - Abigail L Wright
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Kate Devries
- Department of Mining and Materials Engineering, McGill University, Canada
| | - Kerime E Okur
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Michael Jones
- Cell Guidance Systems Ltd, Maia Building, Babraham Bioscience Campus, Cambridge, UK
| | - Imran Masood
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, UK
| | - Lisa J Hill
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, UK
| | - Showan N Nazhat
- Department of Mining and Materials Engineering, McGill University, Canada
| | - Liam M Grover
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Alicia J El Haj
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - Anthony D Metcalfe
- Healthcare Technologies Institute, School of Chemical Engineering, University of Birmingham, Birmingham, UK
| |
Collapse
|
10
|
Santos M, Michael PL, Mitchell TC, Lam YT, Robinson TM, Moore MJ, Tan RP, Rnjak-Kovacina J, Lim KS, Wise SG. On-Demand Bioactivation of Inert Materials With Plasma-Polymerized Nanoparticles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311313. [PMID: 38483292 DOI: 10.1002/adma.202311313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/05/2024] [Indexed: 03/22/2024]
Abstract
Conventional gas plasma treatments are crucial for functionalizing materials in biomedical applications, but have limitations hindering their broader use. These methods require exposure to reactive media under vacuum conditions, rendering them unsuitable for substrates that demand aqueous environments, such as proteins and hydrogels. In addition, complex geometries are difficult to treat, necessitating extensive customization for each material and shape. To address these constraints, an innovative approach employing plasma polymer nanoparticles (PPN) as a versatile functionalization tool is proposed. PPN share similarities with traditional plasma polymer coatings (PPC) but offer unique advantages: compatibility with aqueous systems, the ability to modify complex geometries, and availability as off-the-shelf products. Robust immobilization of PPN on various substrates, including synthetic polymers, proteins, and complex hydrogel structures is demonstrated in this study. This results in substantial improvements in surface hydrophilicity. Materials functionalization with arginylglycylaspartic acid (RGD)-loaded PPN significantly enhances cell attachment, spreading, and substrate coverage on inert scaffolds compared to passive RGD coatings. Improved adhesion to complex geometries and subsequent differentiation following growth factor exposure is also demonstrated. This research introduces a novel substrate functionalization approach that mimics the outcomes of plasma coating technology but vastly expands its applicability, promising advancements in biomedical materials and devices.
Collapse
Affiliation(s)
- Miguel Santos
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Praveesuda L Michael
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Timothy C Mitchell
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Yuen Ting Lam
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Thomas M Robinson
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Mathew J Moore
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Richard P Tan
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, 2006, Australia
| | - Khoon S Lim
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Steven G Wise
- School of Medical Sciences, University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
11
|
Baldassarro VA, Alastra G, Cescatti M, Quadalti C, Lorenzini L, Giardino L, Calzà L. SARS-CoV-2-related peptides induce endothelial-to-mesenchymal transition in endothelial capillary cells derived from different body districts: focus on membrane (M) protein. Cell Tissue Res 2024; 397:241-262. [PMID: 38953987 DOI: 10.1007/s00441-024-03900-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/30/2024] [Indexed: 07/04/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), responsible for the COVID-19, may lead to multiple organ dysfunctions and long-term complications. The induction of microvascular dysfunction is regarded as a main player in these pathological processes. To investigate the possible impact of SARS-CoV-2-induced endothelial-to-mesenchymal transition (EndMT) on fibrosis in "long-COVID" syndrome, we used primary cultures of human microvascular cells derived from the lungs, as the main infection target, compared to cells derived from different organs (dermis, heart, kidney, liver, brain) and to the HUVEC cell line. To mimic the virus action, we used mixed SARS-CoV-2 peptide fragments (PepTivator®) of spike (S), nucleocapsid (N), and membrane (M) proteins. TGFβ2 and cytokine mix (IL-1β, IL-6, TNFα) were used as positive controls. The percentage of cells positive to mesenchymal and endothelial markers was quantified by high content screening. We demonstrated that S+N+M mix induces irreversible EndMT in all analyzed endothelial cells via the TGFβ pathway, as demonstrated by ApoA1 treatment. We then tested the contribution of single peptides in lung and brain cells, demonstrating that EndMT is triggered by M peptide. This was confirmed by transfection experiment, inducing the endogenous expression of the glycoprotein M in lung-derived cells. In conclusion, we demonstrated that SARS-CoV-2 peptides induce EndMT in microvascular endothelial cells from multiple body districts. The different peptides play different roles in the induction and maintenance of the virus-mediated effects, which are organ-specific. These results corroborate the hypothesis of the SARS-CoV-2-mediated microvascular damage underlying the multiple organ dysfunctions and the long-COVID syndrome.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Ozzano dell'Emilia, Bologna, Italy
- Interdepartmental Centre for Industrial Research in Health Sciences and Technology ICIR-HST, University of Bologna, Bologna, Italy
| | - Giuseppe Alastra
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Ozzano dell'Emilia, Bologna, Italy
| | | | - Corinne Quadalti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Luca Lorenzini
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Ozzano dell'Emilia, Bologna, Italy
- Interdepartmental Centre for Industrial Research in Health Sciences and Technology ICIR-HST, University of Bologna, Bologna, Italy
| | - Luciana Giardino
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Ozzano dell'Emilia, Bologna, Italy
- Interdepartmental Centre for Industrial Research in Health Sciences and Technology ICIR-HST, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Interdepartmental Centre for Industrial Research in Health Sciences and Technology ICIR-HST, University of Bologna, Bologna, Italy.
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy.
| |
Collapse
|
12
|
Chen H, Li YY, Nio K, Tang H. Unveiling the Impact of BMP9 in Liver Diseases: Insights into Pathogenesis and Therapeutic Potential. Biomolecules 2024; 14:1013. [PMID: 39199400 PMCID: PMC11353080 DOI: 10.3390/biom14081013] [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: 07/17/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Bone morphogenetic proteins (BMPs) are a group of growth factors belonging to the transforming growth factor β(TGF-β) family. While initially recognized for their role in bone formation, BMPs have emerged as significant players in liver diseases. Among BMPs with various physiological activities, this comprehensive review aims to delve into the involvement of BMP9 specifically in liver diseases and provide insights into the complex BMP signaling pathway. Through an enhanced understanding of BMP9, we anticipate the discovery of new therapeutic options and potential strategies for managing liver diseases.
Collapse
Affiliation(s)
- Han Chen
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China;
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ying-Yi Li
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa 9208641, Japan;
| | - Kouki Nio
- Department of Gastroenterology, Kanazawa University Hospital, Kanazawa 9208641, Japan;
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China;
- Laboratory of Infectious and Liver Diseases, Institute of Infectious Diseases, West China Hospital of Sichuan University, Chengdu 610041, China
| |
Collapse
|
13
|
Horvat Mercnik M, Schliefsteiner C, Sanchez-Duffhues G, Wadsack C. TGFβ signalling: a nexus between inflammation, placental health and preeclampsia throughout pregnancy. Hum Reprod Update 2024; 30:442-471. [PMID: 38519450 PMCID: PMC11215164 DOI: 10.1093/humupd/dmae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/16/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The placenta is a unique and pivotal organ in reproduction, controlling crucial growth and cell differentiation processes that ensure a successful pregnancy. Placental development is a tightly regulated and dynamic process, in which the transforming growth factor beta (TGFβ) superfamily plays a central role. This family of pleiotropic growth factors is heavily involved in regulating various aspects of reproductive biology, particularly in trophoblast differentiation during the first trimester of pregnancy. TGFβ signalling precisely regulates trophoblast invasion and the cell transition from cytotrophoblasts to extravillous trophoblasts, which is an epithelial-to-mesenchymal transition-like process. Later in pregnancy, TGFβ signalling ensures proper vascularization and angiogenesis in placental endothelial cells. Beyond its role in trophoblasts and endothelial cells, TGFβ signalling contributes to the polarization and function of placental and decidual macrophages by promoting maternal tolerance of the semi-allogeneic foetus. Disturbances in early placental development have been associated with several pregnancy complications, including preeclampsia (PE) which is one of the severe complications. Emerging evidence suggests that TGFβ is involved in the pathogenesis of PE, thereby offering a potential target for intervention in the human placenta. OBJECTIVE AND RATIONALE This comprehensive review aims to explore and elucidate the roles of the major members of the TGFβ superfamily, including TGFβs, bone morphogenetic proteins (BMPs), activins, inhibins, nodals, and growth differentiation factors (GDFs), in the context of placental development and function. The review focusses on their interactions within the major cell types of the placenta, namely trophoblasts, endothelial cells, and immune cells, in both normal pregnancies and pregnancies complicated by PE throughout pregnancy. SEARCH METHODS A literature search was carried out using PubMed and Google Scholar, searching terms: 'TGF signalling preeclampsia', 'pregnancy TGF signalling', 'preeclampsia tgfβ', 'preeclampsia bmp', 'preeclampsia gdf', 'preeclampsia activin', 'endoglin preeclampsia', 'endoglin pregnancy', 'tgfβ signalling pregnancy', 'bmp signalling pregnancy', 'gdf signalling pregnancy', 'activin signalling pregnancy', 'Hofbauer cell tgfβ signalling', 'placental macrophages tgfβ', 'endothelial cells tgfβ', 'endothelium tgfβ signalling', 'trophoblast invasion tgfβ signalling', 'trophoblast invasion Smad', 'trophoblast invasion bmp', 'trophoblast invasion tgfβ', 'tgfβ preeclampsia', 'tgfβ placental development', 'TGFβ placental function', 'endothelial dysfunction preeclampsia tgfβ signalling', 'vascular remodelling placenta TGFβ', 'inflammation pregnancy tgfβ', 'immune response pregnancy tgfβ', 'immune tolerance pregnancy tgfβ', 'TGFβ pregnancy NK cells', 'bmp pregnancy NK cells', 'bmp pregnancy tregs', 'tgfβ pregnancy tregs', 'TGFβ placenta NK cells', 'TGFβ placenta tregs', 'NK cells preeclampsia', 'Tregs preeclampsia'. Only articles published in English until 2023 were used. OUTCOMES A comprehensive understanding of TGFβ signalling and its role in regulating interconnected cell functions of the main placental cell types provides valuable insights into the processes essential for successful placental development and growth of the foetus during pregnancy. By orchestrating trophoblast invasion, vascularization, immune tolerance, and tissue remodelling, TGFβ ligands contribute to the proper functioning of a healthy maternal-foetal interface. However, dysregulation of TGFβ signalling has been implicated in the pathogenesis of PE, where the shallow trophoblast invasion, defective vascular remodelling, decreased uteroplacental perfusion, and endothelial cell and immune dysfunction observed in PE, are all affected by an altered TGFβ signalling. WIDER IMPLICATIONS The dysregulation of TGFβ signalling in PE has important implications for research and clinical practice. Further investigation is required to understand the underlying mechanisms, including the role of different ligands and their regulation under pathophysiological conditions, in order to discover new therapeutic targets. Distinguishing between clinically manifested subtypes of PE and studying TGFβ signalling in different placental cell types holistically is an important first step. To put this knowledge into practice, pre-clinical animal models combined with new technologies are needed. This may also lead to improved human research models and identify potential therapeutic targets, ultimately improving outcomes for affected pregnancies and reducing the burden of PE.
Collapse
Affiliation(s)
| | | | - Gonzalo Sanchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Tissue-Specific BMP Signalling ISPA-HUCA, Oviedo, Spain
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
14
|
Singh A, Bhatt KS, Nguyen HC, Frisbee JC, Singh KK. Endothelial-to-Mesenchymal Transition in Cardiovascular Pathophysiology. Int J Mol Sci 2024; 25:6180. [PMID: 38892367 PMCID: PMC11173124 DOI: 10.3390/ijms25116180] [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: 05/10/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Under different pathophysiological conditions, endothelial cells lose endothelial phenotype and gain mesenchymal cell-like phenotype via a process known as endothelial-to-mesenchymal transition (EndMT). At the molecular level, endothelial cells lose the expression of endothelial cell-specific markers such as CD31/platelet-endothelial cell adhesion molecule, von Willebrand factor, and vascular-endothelial cadherin and gain the expression of mesenchymal cell markers such as α-smooth muscle actin, N-cadherin, vimentin, fibroblast specific protein-1, and collagens. EndMT is induced by numerous different pathways triggered and modulated by multiple different and often redundant mechanisms in a context-dependent manner depending on the pathophysiological status of the cell. EndMT plays an essential role in embryonic development, particularly in atrioventricular valve development; however, EndMT is also implicated in the pathogenesis of several genetically determined and acquired diseases, including malignant, cardiovascular, inflammatory, and fibrotic disorders. Among cardiovascular diseases, aberrant EndMT is reported in atherosclerosis, pulmonary hypertension, valvular disease, fibroelastosis, and cardiac fibrosis. Accordingly, understanding the mechanisms behind the cause and/or effect of EndMT to eventually target EndMT appears to be a promising strategy for treating aberrant EndMT-associated diseases. However, this approach is limited by a lack of precise functional and molecular pathways, causes and/or effects, and a lack of robust animal models and human data about EndMT in different diseases. Here, we review different mechanisms in EndMT and the role of EndMT in various cardiovascular diseases.
Collapse
Affiliation(s)
- Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Kriti S. Bhatt
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Hien C. Nguyen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Jefferson C. Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
| | - Krishna K. Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (A.S.); (K.S.B.); (H.C.N.); (J.C.F.)
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
15
|
Vuong CK, Fukushige M, Ngo NH, Yamashita T, Obata-Yasuoka M, Hamada H, Osaka M, Tsukada T, Hiramatsu Y, Ohneda O. Extracellular Vesicles Derived from Type 2 Diabetic Mesenchymal Stem Cells Induce Endothelial Mesenchymal Transition under High Glucose Conditions Through the TGFβ/Smad3 Signaling Pathway. Stem Cells Dev 2024; 33:262-275. [PMID: 38717965 DOI: 10.1089/scd.2023.0262] [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] [Indexed: 06/06/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with endothelial dysfunction, which results in delayed wound healing. Mesenchymal stem cells (MSCs) play a vital role in supporting endothelial cells (ECs) and promoting wound healing by paracrine effects through their secretome-containing extracellular vesicles. We previously reported the impaired wound healing ability of adipose tissue-derived MSC from T2DM donors; however, whether extracellular vesicles isolated from T2DM adipose tissue-derived MSCs (dEVs) exhibit altered functions in comparison to those derived from healthy donors (nEVs) is still unclear. In this study, we found that nEVs induced EC survival and angiogenesis, whereas dEVs lost these abilities. In addition, under high glucose conditions, nEV protected ECs from endothelial-mesenchymal transition (EndMT), whereas dEV significantly induced EndMT by activating the transforming growth factor-β/Smad3 signaling pathway, which impaired the tube formation and in vivo wound healing abilities of ECs. Interestingly, the treatment of dEV-internalized ECs with nEVs rescued the induced EndMT effects. Of note, the internalization of nEV into T2DM adipose tissue-derived MSC resulted in the production of an altered n-dEV, which inhibited EndMT and supported the survival of T2DM db/db mice from severe wounds. Taken together, our findings suggest the role of dEV in endothelial dysfunction and delayed wound healing in T2DM by the promotion of EndMT. Moreover, nEV treatment can be considered a promising candidate for cell-free therapy to protect ECs in T2DM.
Collapse
Affiliation(s)
- Cat-Khanh Vuong
- Graduate School of Comprehensive Human Science, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Mizuho Fukushige
- Graduate School of Comprehensive Human Science, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | - Nhat-Hoang Ngo
- Graduate School of Comprehensive Human Science, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
- PhD Program in Human Biology, University of Tsukuba, Tsukuba, Japan
| | - Toshiharu Yamashita
- Graduate School of Comprehensive Human Science, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| | | | - Hiromi Hamada
- Department of Obstetrics and Gynecology, University of Tsukuba
| | - Motoo Osaka
- Department of Cardiovascular Surgery, University of Tsukuba, Tsukuba, Japan
| | - Toru Tsukada
- Department of Cardiovascular Surgery, University of Tsukuba, Tsukuba, Japan
| | - Yuji Hiramatsu
- Department of Cardiovascular Surgery, University of Tsukuba, Tsukuba, Japan
| | - Osamu Ohneda
- Graduate School of Comprehensive Human Science, Laboratory of Regenerative Medicine and Stem Cell Biology, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
16
|
Feng J, Li K, Xie F, Han L, Wu Y. IL-35 ameliorates lipopolysaccharide-induced endothelial dysfunction by inhibiting endothelial-to-mesenchymal transition. Int Immunopharmacol 2024; 129:111567. [PMID: 38335651 DOI: 10.1016/j.intimp.2024.111567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/12/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome (SIRS) caused mainly by bacterial infection. The morbidity and mortality rates of sepsis are extremely high. About 18 million people worldwide suffer from severe sepsis each year, and about 14,000 people die from it every day. Previous studies have revealed that endothelial dysfunction plays a vital role in the pathological change of sepsis. Furthermore, endothelial-mesenchymal transition (EndMT, EndoMT) is capable of triggering endothelial dysfunction. And yet, it remains obscure whether interleukin-35 (IL-35) can alleviate endothelial dysfunction by attenuating LPS-induced EndMT. Here, through in vivo and in vitro experiments, we revealed that IL-35 has a previously unknown function to attenuate LPS-induced endothelial dysfunction by inhibiting LPS-induced EndMT. Mechanistically, IL-35 acts by regulating the NFκB signaling pathway.
Collapse
Affiliation(s)
- Jie Feng
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Kai Li
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Feng Xie
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
17
|
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
|
18
|
Xu X, Zhang B, Wang Y, Shi S, Lv J, Fu Z, Gao X, Li Y, Wu H, Song Q. Renal fibrosis in type 2 cardiorenal syndrome: An update on mechanisms and therapeutic opportunities. Biomed Pharmacother 2023; 164:114901. [PMID: 37224755 DOI: 10.1016/j.biopha.2023.114901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023] Open
Abstract
Cardiorenal syndrome (CRS) is a state of coexisting heart failure and renal insufficiency in which acute or chronic dysfunction of the heart or kidney lead to acute or chronic dysfunction of the other organ.It was found that renal fibrosis is an important pathological process in the progression of type 2 CRS to end-stage renal disease, and progressive renal impairment accelerates the deterioration of cardiac function and significantly increases the hospitalization and mortality rates of patients. Previous studies have found that Hemodynamic Aiteration, RAAS Overactivation, SNS Dysfunction, Endothelial Dysfunction and Imbalance of natriuretic peptide system contribute to the development of renal disease in the decompensated phase of heart failure, but the exact mechanisms is not clear. Therefore, in this review, we focus on the molecular pathways involved in the development of renal fibrosis due to heart failure and identify the canonical and non-canonical TGF-β signaling pathways and hypoxia-sensing pathways, oxidative stress, endoplasmic reticulum stress, pro-inflammatory cytokines and chemokines as important triggers and regulators of fibrosis development, and summarize the therapeutic approaches for the above signaling pathways, including SB-525334 Sfrp1, DKK1, IMC, rosarostat, 4-PBA, etc. In addition, some potential natural drugs for this disease are also summarized, including SQD4S2, Wogonin, Astragaloside, etc.
Collapse
Affiliation(s)
- Xia Xu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajiao Wang
- College of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Shuqing Shi
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayu Lv
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xiya Gao
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Huaqin Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
19
|
Yang X, Cheng K, Wang LY, Jiang JG. The role of endothelial cell in cardiac hypertrophy: Focusing on angiogenesis and intercellular crosstalk. Biomed Pharmacother 2023; 163:114799. [PMID: 37121147 DOI: 10.1016/j.biopha.2023.114799] [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: 03/01/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/02/2023] Open
Abstract
Cardiac hypertrophy is characterized by cardiac structural remodeling, fibrosis, microvascular rarefaction, and chronic inflammation. The heart is structurally organized by different cell types, including cardiomyocytes, fibroblasts, endothelial cells, and immune cells. These cells highly interact with each other by a number of paracrine or autocrine factors. Cell-cell communication is indispensable for cardiac development, but also plays a vital role in regulating cardiac response to damage. Although cardiomyocytes and fibroblasts are deemed as key regulators of hypertrophic stimulation, other cells, including endothelial cells, also exert important effects on cardiac hypertrophy. More particularly, endothelial cells are the most abundant cells in the heart, which make up the basic structure of blood vessels and are widespread around other cells in the heart, implicating the great and inbuilt advantage of intercellular crosstalk. Cardiac microvascular plexuses are essential for transport of liquids, nutrients, molecules and cells within the heart. Meanwhile, endothelial cell-mediated paracrine signals have multiple positive or negative influences on cardiac hypertrophy. However, a comprehensive discussion of these influences and consequences is required. This review aims to summarize the basic function of endothelial cells in angiogenesis, with an emphasis on angiogenic molecules under hypertrophic conditions. The secondary objective of the research is to fully discuss the key molecules involved in the intercellular crosstalk and the endothelial cell-mediated protective or detrimental effects on other cardiac cells. This review provides a more comprehensive understanding of the overall role of endothelial cells in cardiac hypertrophy and guides the therapeutic approaches and drug development of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xing Yang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China
| | - Kun Cheng
- Hepatic Surgery Centre, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430000, China
| | - Lu-Yun Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| | - Jian-Gang Jiang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430000, China.
| |
Collapse
|
20
|
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
|
21
|
Weber CM, Harris MN, Zic SM, Sangha GS, Arnold NS, Dluzen DF, Clyne AM. Angiotensin II Increases Oxidative Stress and Inflammation in Female, But Not Male, Endothelial Cells. Cell Mol Bioeng 2023; 16:127-141. [PMID: 37096068 PMCID: PMC10121986 DOI: 10.1007/s12195-023-00762-2] [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: 10/19/2022] [Accepted: 03/29/2023] [Indexed: 04/26/2023] Open
Abstract
Introduction Women are at elevated risk for certain cardiovascular diseases, including pulmonary arterial hypertension, Alzheimer's disease, and vascular complications of diabetes. Angiotensin II (AngII), a circulating stress hormone, is elevated in cardiovascular disease; however, our knowledge of sex differences in the vascular effects of AngII are limited. We therefore analyzed sex differences in human endothelial cell response to AngII treatment. Methods Male and female endothelial cells were treated with AngII for 24 h and analyzed by RNA sequencing. We then used endothelial and mesenchymal markers, inflammation assays, and oxidative stress indicators to measure female and male endothelial cell functional changes in response to AngII. Results Our data show that female and male endothelial cells are transcriptomically distinct. Female endothelial cells treated with AngII had widespread gene expression changes related to inflammatory and oxidative stress pathways, while male endothelial cells had few gene expression changes. While both female and male endothelial cells maintained their endothelial phenotype with AngII treatment, female endothelial cells showed increased release of the inflammatory cytokine interleukin-6 and increased white blood cell adhesion following AngII treatment concurrent with a second inflammatory cytokine. Additionally, female endothelial cells had elevated reactive oxygen species production compared to male endothelial cells after AngII treatment, which may be partially due to nicotinamide adenine dinucleotide phosphate oxidase-2 (NOX2) escape from X-chromosome inactivation. Conclusions These data suggest that endothelial cells have sexually dimorphic responses to AngII, which could contribute to increased prevalence of some cardiovascular diseases in women. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-023-00762-2.
Collapse
Affiliation(s)
- Callie M. Weber
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| | - Mikayla N. Harris
- Department of Biology, Morgan State University, Baltimore, MD 21251 USA
| | - Sophia M. Zic
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| | - Gurneet S. Sangha
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| | - Nicole S. Arnold
- Department of Biology, Morgan State University, Baltimore, MD 21251 USA
| | - Douglas F. Dluzen
- Department of Biology, Morgan State University, Baltimore, MD 21251 USA
| | - Alisa Morss Clyne
- Fischell Department of Bioengineering, University of Maryland, 8278 Paint Branch Dr., College Park, MD 20742 USA
| |
Collapse
|
22
|
Zamani M, Cheng YH, Charbonier F, Gupta VK, Mayer AT, Trevino AE, Quertermous T, Chaudhuri O, Cahan P, Huang NF. Single-Cell Transcriptomic Census of Endothelial Changes Induced by Matrix Stiffness and the Association with Atherosclerosis. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2203069. [PMID: 36816792 PMCID: PMC9937733 DOI: 10.1002/adfm.202203069] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Indexed: 05/28/2023]
Abstract
Vascular endothelial cell (EC) plasticity plays a critical role in the progression of atherosclerosis by giving rise to mesenchymal phenotypes in the plaque lesion. Despite the evidence for arterial stiffening as a major contributor to atherosclerosis, the complex interplay among atherogenic stimuli in vivo has hindered attempts to determine the effects of extracellular matrix (ECM) stiffness on endothelial-mesenchymal transition (EndMT). To study the regulatory effects of ECM stiffness on EndMT, an in vitro model is developed in which human coronary artery ECs are cultured on physiological or pathological stiffness substrates. Leveraging single-cell RNA sequencing, cell clusters with mesenchymal transcriptional features are identified to be more prevalent on pathological substrates than physiological substrates. Trajectory inference analyses reveal a novel mesenchymal-to-endothelial reverse transition, which is blocked by pathological stiffness substrates, in addition to the expected EndMT trajectory. ECs pushed to a mesenchymal character by pathological stiffness substrates are enriched in transcriptional signatures of atherosclerotic ECs from human and murine plaques. This study characterizes at single-cell resolution the transcriptional programs that underpin EC plasticity in both physiological or pathological milieus, and thus serves as a valuable resource for more precisely defining EndMT and the transcriptional programs contributing to atherosclerosis.
Collapse
Affiliation(s)
- Maedeh Zamani
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
| | - Yu-Hao Cheng
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Frank Charbonier
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Vivek Kumar Gupta
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | | | | | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Patrick Cahan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
23
|
Mendez PL, Obendorf L, Jatzlau J, Burdzinski W, Reichenbach M, Nageswaran V, Haghikia A, Stangl V, Hiepen C, Knaus P. Atheroprone fluid shear stress-regulated ALK1-Endoglin-SMAD signaling originates from early endosomes. BMC Biol 2022; 20:210. [PMID: 36171573 PMCID: PMC9520843 DOI: 10.1186/s12915-022-01396-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fluid shear stress enhances endothelial SMAD1/5 signaling via the BMP9-bound ALK1 receptor complex supported by the co-receptor Endoglin. While moderate SMAD1/5 activation is required to maintain endothelial quiescence, excessive SMAD1/5 signaling promotes endothelial dysfunction. Increased BMP signaling participates in endothelial-to-mesenchymal transition and inflammation culminating in vascular diseases such as atherosclerosis. While the function of Endoglin has so far been described under picomolar concentrations of BMP9 and short-term shear application, we investigated Endoglin under physiological BMP9 and long-term pathophysiological shear conditions. RESULTS We report here that knock-down of Endoglin leads to exacerbated SMAD1/5 phosphorylation and atheroprone gene expression profile in HUVECs sheared for 24 h. Making use of the ligand-trap ALK1-Fc, we furthermore show that this increase is dependent on BMP9/10. Mechanistically, we reveal that long-term exposure of ECs to low laminar shear stress leads to enhanced Endoglin expression and endocytosis of Endoglin in Caveolin-1-positive early endosomes. In these endosomes, we could localize the ALK1-Endoglin complex, labeled BMP9 as well as SMAD1, highlighting Caveolin-1 vesicles as a SMAD signaling compartment in cells exposed to low atheroprone laminar shear stress. CONCLUSIONS We identified Endoglin to be essential in preventing excessive activation of SMAD1/5 under physiological flow conditions and Caveolin-1-positive early endosomes as a new flow-regulated signaling compartment for BMP9-ALK1-Endoglin signaling axis in atheroprone flow conditions.
Collapse
Affiliation(s)
- Paul-Lennard Mendez
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- International Max-Planck Research School for Biology and Computation, Berlin, Germany
| | - Leon Obendorf
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Jerome Jatzlau
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Wiktor Burdzinski
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Berlin School for Regenerative Therapies, Berlin, Germany
| | - Maria Reichenbach
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
| | - Vanasa Nageswaran
- Charité-Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Arash Haghikia
- Charité-Universitätsmedizin Berlin, Klinik für Kardiologie, Campus Benjamin Franklin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany
| | - Verena Stangl
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany
- Faculty of Engineering and Natural Sciences, Westphalian University of Applied Sciences, Recklinghausen, Germany
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Berlin, Germany.
- International Max-Planck Research School for Biology and Computation, Berlin, Germany.
- Berlin School for Regenerative Therapies, Berlin, Germany.
| |
Collapse
|
24
|
Chan MKK, Chung JYF, Tang PCT, Chan ASW, Ho JYY, Lin TPT, Chen J, Leung KT, To KF, Lan HY, Tang PMK. TGF-β signaling networks in the tumor microenvironment. Cancer Lett 2022; 550:215925. [DOI: 10.1016/j.canlet.2022.215925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/05/2022] [Accepted: 09/17/2022] [Indexed: 11/02/2022]
|
25
|
He J, Hu W, Ouyang Q, Zhang S, He L, Chen W, Li X, Hu C. Helicobacter pylori infection induces stem cell-like properties in Correa cascade of gastric cancer. Cancer Lett 2022; 542:215764. [PMID: 35654291 DOI: 10.1016/j.canlet.2022.215764] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 02/09/2023]
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer-related death. Its poor prognosis is attributed to unclear pathogenesis. Currently, the most widely accepted model for elucidating the mechanism of GC is the Correa cascade, which covers several histological lesions of the gastric mucosa. GC stem cells (CSCs) are crucial for oncogenesis in the Correa cascade and GC progression. As Helicobacter pylori (H. pylori) is the etiological factor in the Correa cascade, growing evidence suggests that enhancement of gastric stem cell-like properties and increase in CSCs correlate with H. pylori infection. In this paper, we review recent studies that present pathogenic mechanisms by which H. pylori induces gastric stem cell-like properties and CSCs, which may supplement the existing Correa model of GC. First, the dysfunction of developmental signaling pathways associated with H. pylori infection leads to the enhancement of gastric stemness. Second, H. pylori infection promotes alteration of the gastric mucosal microenvironment. In addition, epithelial-mesenchymal transition (EMT) may contribute to H. pylori-induced gastric stemness. Taken together, understanding these pathogeneses will provide potential therapeutic targets for the treatment of CSCs and malignant GC in H. pylori induced-Correa cascade of GC.
Collapse
Affiliation(s)
- JunJian He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - WeiChao Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Army Medical University, Chongqing, 400038, China
| | - ShengWei Zhang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - LiJiao He
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - WeiYan Chen
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - XinZhe Li
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| | - ChangJiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China.
| |
Collapse
|
26
|
Liu F, Chen S, Yu Y, Huang C, Chen H, Wang L, Zhang W, Wu J, Ye Y. Inhibitor of DNA binding 2 knockdown inhibits the growth and liver metastasis of colorectal cancer. Gene 2022; 819:146240. [PMID: 35114275 DOI: 10.1016/j.gene.2022.146240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Liver metastasis of colorectal cancer (CRC) remains high mortality and the mechanism is still unknown. Here we investigated the effects of inhibitor of DNA binding 2 (Id2) on growth and liver metastasis of CRC. METHODS qPCR and western blotting were used to demonstrate mRNA and protein expressions in Id2-knockdown HCT116 cells. Cell growth was observed by cell proliferation assay, colony formation assay and flow cytometry. Cell migration and invasion were observed with wound healing assay and transwell migration and invasion assay. The effects of Id2 knockdown on tumor growth and liver metastasis in vivo were evaluated respectively with subcutaneous tumor model and colorectal liver metastasis model by injecting HCT116 cells into the mesentery triangle of cecum in mice. RESULTS Id2 overexpression was found in CRC cell lines. Id2 knockdown resulted in a reduction in the proliferation, colony formation, migration and invasion of HCT116 cells. The suppression of cell proliferation was accompanied by the cell cycle arrest in the G0/G1 phase with down-regulation of Cyclin D1, Cyclin E, p-Cdk2/3, Cdk6, p-p27 and up-regulation of p21 and p27. Id2 knockdown reversed epithelial-mesenchymal transition (EMT) through increasing E-Cadherin and inhibiting N-Cadherin, Vimentin, β-catenin, Snail and Slug. Id2 was also found to inhibit CRC metastasis via MMP2, MMP9 and TIMP-1. Furthermore, Id2 knockdown suppressed CRC liver metastasis in vivo. CONCLUSION Id2 promotes CRC growth through activation of the PI3K/AKT signaling pathway, and triggers EMT to enhance CRC migration and invasion.
Collapse
Affiliation(s)
- Fang Liu
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Shuping Chen
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Yue Yu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Chuanzhong Huang
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Huijing Chen
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Ling Wang
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China
| | - Wanping Zhang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Junxin Wu
- Department of Radiation Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China.
| | - Yunbin Ye
- Laboratory of Immuno-Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou 350014, China; The School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou 350014, China.
| |
Collapse
|
27
|
Chen W, Gard JMC, Epshtein Y, Camp SM, Garcia JGN, Jacobson JR, Cress AE. Integrin Beta 4E Promotes Endothelial Phenotypic Changes and Attenuates Lung Endothelial Cell Inflammatory Responses. Front Physiol 2022; 13:769325. [PMID: 35250607 PMCID: PMC8895044 DOI: 10.3389/fphys.2022.769325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
We previously reported integrin beta 4 (ITGB4) is an important mediator of lung vascular protection by simvastatin, a 3-hydroxy-3-methylglutaryl-coenzyme A-reductase inhibitor. In this study, we report increased endothelial cell (EC) expression specifically of ITGB4E, an ITGB4 mRNA splice variant, by simvastatin with effects on EC protein expression and inflammatory responses. In initial experiments, human pulmonary artery ECs were treated using simvastatin (5 μM, 24 h) prior to immunoprecipitation of integrin alpha 6 (ITGA6), which associates with ITGB4, and Western blotting for full-length ITGB4 and ITGB4E, uniquely characterized by a truncated 114 amino acid cytoplasmic domain. These experiments confirmed a significant increase in both full-length ITGB4 and ITGB4E. To investigate the effects of increased ITGB4E expression alone, ECs were transfected with ITGB4E or control vector, and cells were seeded in wells containing Matrigel to assess effects on angiogenesis or used for scratch assay to assess migration. Decreased angiogenesis and migration were observed in ITGB4E transfected ECs compared with controls. In separate experiments, PCR and Western blots from transfected cells demonstrated significant changes in EC protein expression associated with increased ITGB4E, including marked decreases in platelet endothelial cell adhesion molecule-1 (PECAM-1) and vascular endothelial-cadherin (VE-cadherin) as well as increased expression of E-cadherin and N-cadherin along with increased expression of the Slug and Snail transcription factors that promote endothelial-to-mesenchymal transition (EndMT). We, then, investigated the functional effects of ITGB4E overexpression on EC inflammatory responses and observed a significant attenuation of lipopolysaccharide (LPS)-induced mitogen-activated protein kinase (MAPK) activation, including decreased phosphorylation of both extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK), as well as reduced inflammatory cytokines (IL-6 and IL-8), expressed in the media of EC after either LPS or excessive cyclic stretch (CS). Finally, EC expression-increased ITGB4E demonstrated decreased barrier disruption induced by thrombin as measured by transendothelial electrical resistance. Our data support distinct EC phenotypic changes induced by ITGB4E that are also associated with an attenuation of cellular inflammatory responses. These findings implicate ITGB4E upregulation as an important mediator of lung EC protection by statins and may lead to novel therapeutic strategies for patients with or at risk for acute lung injury (ALI).
Collapse
Affiliation(s)
- Weiguo Chen
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Jamie M. C. Gard
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| | - Yulia Epshtein
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
| | - Sara M. Camp
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Joe G. N. Garcia
- Department of Medicine, University of Arizona Health Sciences, Tucson, AZ, United States
| | - Jeffrey R. Jacobson
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Jeffrey R. Jacobson,
| | - Anne E. Cress
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
28
|
Li Z, Duan H, Jia Y, Zhao C, Li W, Wang X, Gong Y, Dong C, Ma B, Dou S, Zhang B, Li D, Cao Y, Xie L, Zhou Q, Shi W. Long-term corneal recovery by simultaneous delivery of hPSC-derived corneal endothelial precursors and nicotinamide. J Clin Invest 2022; 132:146658. [PMID: 34981789 PMCID: PMC8718141 DOI: 10.1172/jci146658] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 11/02/2021] [Indexed: 12/12/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) hold great promise for the treatment of various human diseases. However, their therapeutic benefits and mechanisms for treating corneal endothelial dysfunction remain undefined. Here, we developed a therapeutic regimen consisting of the combination of hPSC-derived corneal endothelial precursors (CEPs) with nicotinamide (NAM) for effective treatment of corneal endothelial dysfunction. In rabbit and nonhuman primate models, intracameral injection of CEPs and NAM achieved long-term recovery of corneal clarity and thickness, similar with the therapeutic outcome of cultured human corneal endothelial cells (CECs). The transplanted human CEPs exhibited structural and functional integration with host resident CECs. However, the long-term recovery relied on the stimulation of endogenous endothelial regeneration in rabbits, but predominantly on the replacing function of transplanted cells during the 3-year follow-up in nonhuman primates, which resemble human corneal endothelium with limited regenerative capacity. Mechanistically, NAM ensured in vivo proper maturation of transplanted CEPs into functional CECs by preventing premature senescence and endothelial-mesenchymal transition within the TGF-β–enriched aqueous humor. Together, we provide compelling experimental evidence and mechanistic insights of simultaneous delivery of CEPs and NAM as a potential approach for treating corneal endothelial dysfunction.
Collapse
Affiliation(s)
- Zongyi Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Haoyun Duan
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Yanni Jia
- Eye Hospital of Shandong First Medical University, Jinan, China
| | - Can Zhao
- Eye Hospital of Shandong First Medical University, Jinan, China
| | - Wenjing Li
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China
| | - Xin Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Yajie Gong
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Chunxiao Dong
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| | - Bochao Ma
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Bin Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Dongfang Li
- Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Yihai Cao
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Lixin Xie
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Weiyun Shi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China.,Eye Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
29
|
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
|