1
|
Han X, Li Y, Wang E, Zhu H, Huang X, Pu W, Zhang M, Liu K, Zhao H, Liu Z, Zhao Y, Shen L, Li Y, Yang X, Wang QD, Ma X, Shen R, O Lui K, Wang L, He B, Zhou B. Exploring Origin-Dependent Susceptibility of Smooth Muscle Cells to Aortic Diseases Through Intersectional Genetics. Circulation 2025; 151:1248-1267. [PMID: 39925267 DOI: 10.1161/circulationaha.124.070782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND The developmental diversity among smooth muscle cells (SMCs) plays a crucial role in segment-specific aortic diseases. However, traditional genetic approaches are inadequate for enabling in vivo analysis of disease susceptibility associated with cellular origin. There is an urgent need to build genetic technologies that target different developmental origins to investigate the mechanisms of aortopathies, thereby facilitating the development of effective therapeutics. METHODS To address this challenge, we developed an advanced dual recombinase-mediated intersectional genetic system, specifically designed to precisely target SMCs from various developmental origins in mice. Specifically, we used Isl1-Dre, Wnt1-Dre, Meox1-DreER, and Upk3b-Dre to target SMC progenitors from the second heart field, cardiac neural crest, somites, and mesothelium, respectively. This system was combined with single-cell RNA sequencing to investigate the impact of TGF-β (transforming growth factor-β) signaling in different segments of the aorta by selectively knocking out Tgfbr2 in the ascending aorta and Smad4 in the aortic arch, respectively. RESULTS Through intersectional genetic approaches, we use the Myh11-Cre(ER) driver along with origin-specific Dre drivers to trace cells of diverse developmental origins within the SMC population. We found that a deficiency of Tgfbr2 in SMCs of the ascending aorta leads to aneurysm formation in this specific region. We also demonstrate the critical role of Smad4 in preserving aortic wall integrity and homeostasis in SMCs of the aortic arch. CONCLUSIONS Our approach to genetically targeting SMC subtypes provides a novel platform for exploring origin-dependent or location-specific aortic vascular diseases. This genetic system enables comprehensive analysis of contributions from different cell lineages to SMC behavior and pathology, thereby paving the way for targeted research and therapeutic interventions in the future.
Collapse
MESH Headings
- Animals
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Mice
- Receptor, Transforming Growth Factor-beta Type II/genetics
- Receptor, Transforming Growth Factor-beta Type II/metabolism
- Receptor, Transforming Growth Factor-beta Type II/deficiency
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Knockout
- Genetic Predisposition to Disease
- Aorta/metabolism
- Aorta/pathology
- Signal Transduction
Collapse
Affiliation(s)
- Ximeng Han
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Enci Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
| | - Huan Zhu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Xiuzhen Huang
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Wenjuan Pu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Mingjun Zhang
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Kuo Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou (K.L., B.Z.)
| | - Huan Zhao
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Zixin Liu
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
| | - Yufei Zhao
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
| | - Linghong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Yan Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, China (Y.L.)
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China (Y.L.)
| | - Xiao Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing Institute of Lifeomics, China (X.Y.)
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Q.-D.W.)
| | - Xin Ma
- Department of Pharmacology, Wuxi School of Medicine, Jiangnan University, China (X.M.)
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, China (R.S.)
| | - Kathy O Lui
- CAS CEMCS-CUHK Joint Laboratories, Department of Chemical Pathology; Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, Chinese University of Hong Kong, China (K.O.L.)
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China (E.W., Y.Z., L.W.)
- Department of Vascular Surgery (Xiamen), Zhongshan Hospital, Fudan University, Xiamen, China (L.W.)
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, China (X. Han, Y.L., L.S., B.H.)
| | - Bin Zhou
- CAS CEMCS-CUHK Joint Laboratories, New Cornerstone Science Laboratory, Key Laboratory of Multi-Cell Systems, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai (X. Han, H. Zhu, X. Huang, W.P., M.Z., H. Zhao, Z.L., B.Z.)
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou (K.L., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (B.Z.)
| |
Collapse
|
2
|
Paterek A, Załęska-Kocięcka M, Wojdyńska Z, Kalisz M, Litwiniuk A, Leszek P, Mączewski M. Epicardial fat in heart failure-Friend, foe, or bystander. Obes Rev 2024; 25:e13820. [PMID: 39187402 DOI: 10.1111/obr.13820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 07/12/2024] [Accepted: 08/02/2024] [Indexed: 08/28/2024]
Abstract
Epicardial adipose tissue (EAT) is a fat depot covering the heart. No physical barrier separates EAT from the myocardium, so EAT can easily affect the underlying cardiac muscle. EAT can participate in the development and progression of heart failure with preserved (HFpEF) and reduced ejection fraction (HFrEF). In healthy humans, excess EAT is associated with impaired cardiac function and worse outcomes. In HFpEF, this trend continues: EAT amount is usually increased, and excess EAT correlates with worse function/outcomes. However, in HFrEF, the opposite is true: reduced EAT amount correlates with worse cardiac function/outcomes. Surprisingly, although EAT has beneficial effects on cardiac function, it aggravates ventricular arrhythmias. Here, we dissect these phenomena, trying to explain these paradoxical findings to find a target for novel heart failure therapies aimed at EAT rather than the myocardium itself. However, the success of this approach depends on a thorough understanding of interactions between EAT and the myocardium.
Collapse
Affiliation(s)
- Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marta Załęska-Kocięcka
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Zuzanna Wojdyńska
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Małgorzata Kalisz
- Department of Clinical Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Anna Litwiniuk
- Department of Clinical Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Przemysław Leszek
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
3
|
Yang X, He S, Li X, Guo Z, Wang H, Zhang Z, Song X, Jia K, He L, Zhou B. Synchronized lineage tracing of cell membranes and nuclei by dual recombinases and dual fluorescent. J Genet Genomics 2024; 51:1474-1484. [PMID: 38996840 DOI: 10.1016/j.jgg.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
Genetic lineage tracing has been widely employed to investigate cell lineages and fate. However, conventional reporting systems often label the entire cytoplasm, making it challenging to discern cell boundaries. Additionally, single Cre-loxP recombination systems have limitations in tracing specific cell populations. This study proposes three reporting systems utilizing Cre, Dre, and Dre+Cre mediated recombination. These systems incorporate tdTomato expression on the cell membrane and PhiYFP expression within the nucleus, allowing for clear observation of the nucleus and membrane. The efficacy of these systems is successfully demonstrated by labeling cardiomyocytes and hepatocytes. The potential for dynamic visualization of the cell membrane is showcased using intravital imaging microscopy or three-dimensional imaging. Furthermore, by combining this dual recombinase system with the ProTracer system, hepatocyte proliferation is traced with enhanced precision. This reporting system holds significant importance for advancing the understanding of cell fate studies in development, homeostasis, and diseases.
Collapse
Affiliation(s)
- Xueying Yang
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Shun He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Xufeng Li
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China
| | - Zhihou Guo
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Haichang Wang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Zhuonan Zhang
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Xin Song
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Ke Jia
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China.
| | - Bin Zhou
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China; New Cornerstone Science Laboratory, State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
4
|
Nogajski Ł, Mazuruk M, Kacperska M, Kurpias M, Mączewski M, Nowakowski M, Mączewski M, Michałowska I, Leszek P, Paterek A. Epicardial fat density obtained with computed tomography imaging - more important than volume? Cardiovasc Diabetol 2024; 23:389. [PMID: 39472958 PMCID: PMC11523889 DOI: 10.1186/s12933-024-02474-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024] Open
Abstract
Epicardial adipose tissue (EAT) is a unique fat depot located between the myocardium and the visceral layer of pericardium. It can be further subdivided into pericoronary (PCAT), periatrial (PAAT) and periventricular adipose tissue (PVentAT), each of them exhibiting specific characteristics and association with the underlying tissue. Since no physical barrier separates EAT from the myocardium, this fat tissue can easily interact with the underlying cardiac structure. EAT can be visualized using various imaging modalities. Computed tomography provides not only information on EAT volume, but also on its density. Indeed, EAT density reflected by the recently developed fat attenuation index (FAI) is emerging as a useful index of PCAT inflammation, PAAT inflammation and fibrosis, while the relevance of density of PVentAT is much less known. The emerging data indicates that FAI can be an important diagnostic and prognostic tool in both coronary artery disease and atrial fibrillation. Future studies will demonstrate if it also could be used as a marker of efficacy of therapies and whether FAI PVentAT could indicate ventricular pathologies, such as heart failure. The aim of the review is to present computed tomography derived FAI as an important tool both to study and better understand the epicardial fat and as a potential predictive marker in cardiovascular disorders.
Collapse
Affiliation(s)
- Łukasz Nogajski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maciej Mazuruk
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Marta Kacperska
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Mikołaj Kurpias
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maciej Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Maksymilian Nowakowski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
- Student's Cardiovascular Scientific Club "Kardioplegia", Medical University of Warsaw, Warsaw, Poland
| | - Michał Mączewski
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Ilona Michałowska
- Department of Radiology, National Institute of Cardiology, Warsaw, Poland
| | - Przemysław Leszek
- Heart Failure and Transplantology Department, Mechanical Circulatory Support and Transplant Department, National Institute of Cardiology, Warsaw, Poland
| | - Aleksandra Paterek
- Department of Clinical Physiology, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
5
|
Cai X, Han M, Lou F, Sun Y, Yin Q, Sun L, Wang Z, Li X, Zhou H, Xu Z, Wang H, Deng S, Zheng X, Zhang T, Li Q, Zhou B, Wang H. Tenascin C + papillary fibroblasts facilitate neuro-immune interaction in a mouse model of psoriasis. Nat Commun 2023; 14:2004. [PMID: 37037861 PMCID: PMC10086024 DOI: 10.1038/s41467-023-37798-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 03/29/2023] [Indexed: 04/12/2023] Open
Abstract
Dermal fibroblasts and cutaneous nerves are important players in skin diseases, while their reciprocal roles during skin inflammation have not been characterized. Here we identify an inflammation-induced subset of papillary fibroblasts that promotes aberrant neurite outgrowth and psoriasiform skin inflammation by secreting the extracellular matrix protein tenascin-C (TNC). Single-cell analysis of fibroblast lineages reveals a Tnc+ papillary fibroblast subset with pro-axonogenesis and neuro-regulation transcriptomic hallmarks. TNC overexpression in fibroblasts boosts neurite outgrowth in co-cultured neurons, while fibroblast-specific TNC ablation suppresses hyperinnervation and alleviates skin inflammation in male mice modeling psoriasis. Dermal γδT cells, the main producers of type 17 pathogenic cytokines, frequently contact nerve fibers in mouse psoriasiform lesions and are likely modulated by postsynaptic signals. Overall, our results highlight the role of an inflammation-responsive fibroblast subset in facilitating neuro-immune synapse formation and suggest potential avenues for future therapeutic research.
Collapse
Affiliation(s)
- Xiaojie Cai
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Maoying Han
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fangzhou Lou
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Yang Sun
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Qianqian Yin
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Libo Sun
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Zhikai Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiangxiao Li
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Hong Zhou
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Zhenyao Xu
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Hong Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Siyu Deng
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Xichen Zheng
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Taiyu Zhang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Qun Li
- The Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Honglin Wang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China.
| |
Collapse
|
6
|
Han M, Liu Z, Liu L, Huang X, Wang H, Pu W, Wang E, Liu X, Li Y, He L, Li X, Wu J, Qiu L, Shen R, Wang QD, Ji Y, Ardehali R, Shu Q, Lui KO, Wang L, Zhou B. Dual genetic tracing reveals a unique fibroblast subpopulation modulating cardiac fibrosis. Nat Genet 2023; 55:665-678. [PMID: 36959363 DOI: 10.1038/s41588-023-01337-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 02/15/2023] [Indexed: 03/25/2023]
Abstract
After severe heart injury, fibroblasts are activated and proliferate excessively to form scarring, leading to decreased cardiac function and eventually heart failure. It is unknown, however, whether cardiac fibroblasts are heterogeneous with respect to their degree of activation, proliferation and function during cardiac fibrosis. Here, using dual recombinase-mediated genetic lineage tracing, we find that endocardium-derived fibroblasts preferentially proliferate and expand in response to pressure overload. Fibroblast-specific proliferation tracing revealed highly regional expansion of activated fibroblasts after injury, whose pattern mirrors that of endocardium-derived fibroblast distribution in the heart. Specific ablation of endocardium-derived fibroblasts alleviates cardiac fibrosis and reduces the decline of heart function after pressure overload injury. Mechanistically, Wnt signaling promotes activation and expansion of endocardium-derived fibroblasts during cardiac remodeling. Our study identifies endocardium-derived fibroblasts as a key fibroblast subpopulation accounting for severe cardiac fibrosis after pressure overload injury and as a potential therapeutic target against cardiac fibrosis.
Collapse
Affiliation(s)
- Maoying Han
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Zixin Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
| | - Lei Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiuzhen Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
| | - Haixiao Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
| | - Wenjuan Pu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
| | - Enci Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiuxiu Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Xufeng Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, China
| | - Jiayu Wu
- Chinese Aacademy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Lin Qiu
- Chinese Aacademy of Sciences Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai, China
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yong Ji
- The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Reza Ardehali
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Qiang Shu
- Department of Pediatric Cardiaovascular Surgery, Children's Hospital, School of Medicine, Zhejiang University, National Clinical Research Center for Child Health, Hangzhou, China
| | - Kathy O Lui
- Department of Chemical Pathology and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Lixin Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Vascular Surgery, Zhongshan Hospital (Xiamen), Fudan University, Xiamen, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences University of the Chinese Academy of Sciences, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, China.
- The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.
- New Cornerstone Science Laboratory, Shenzhen, China.
| |
Collapse
|
7
|
Genetic lineage tracing identifies cardiac mesenchymal-to-adipose transition in an arrhythmogenic cardiomyopathy model. SCIENCE CHINA. LIFE SCIENCES 2023; 66:51-66. [PMID: 36322324 DOI: 10.1007/s11427-022-2176-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is one of the most common inherited cardiomyopathies, characterized by progressive fibrofatty replacement in the myocardium. However, the cellular origin of cardiac adipocytes in ACM remains largely unknown. Unraveling the cellular source of cardiac adipocytes in ACM would elucidate the underlying pathological process and provide a potential target for therapy. Herein, we generated an ACM mouse model by inactivating desmosomal gene desmoplakin in cardiomyocytes; and examined the adipogenic fates of several cell types in the disease model. The results showed that SOX9+, PDGFRa+, and PDGFRb+ mesenchymal cells, but not cardiomyocytes or smooth muscle cells, contribute to the intramyocardial adipocytes in the ACM model. Mechanistically, Bmp4 was highly expressed in the ACM mouse heart and functionally promoted cardiac mesenchymal-to-adipose transition in vitro.
Collapse
|
8
|
Zhang Z, Zhou B. Generation of Plvap-CreER and Car4-CreER for genetic targeting of distinct lung capillary populations. J Genet Genomics 2022; 49:1093-1100. [PMID: 36028133 DOI: 10.1016/j.jgg.2022.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 01/14/2023]
Abstract
It has been reported recently that there are two distinct subpopulations of capillary endothelial cells in the mammalian lungs: gCap (general capillary) and aCap (aerocyte). They are identified by two unique markers, respectively: plasmalemmal vesicle-associated protein (PLVAP) and carbonic anhydrase IV (CAR4). Here, we report two novel knock-in mouse lines Plvap-CreER and Car4-CreER, which genetically target gCap and aCap, respectively. Induced by tamoxifen, the Plvap-CreER and Car4-CreER alleles mediate specific and efficient Cre-loxP recombinations in PLVAP+ gCap and CAR4+ aCap, respectively, in the lungs. These two mouse lines are useful genetic tools to investigate cell fates and functions of PLVAP+ and CAR4+ cells in lung homeostasis, injury and repair.
Collapse
Affiliation(s)
- Zhenqian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
9
|
Liu K, Jin H, Tang M, Zhang S, Tian X, Zhang M, Han X, Liu X, Tang J, Pu W, Li Y, He L, Yang Z, Lui KO, Zhou B. Lineage tracing clarifies the cellular origin of tissue-resident macrophages in the developing heart. J Biophys Biochem Cytol 2022; 221:213182. [PMID: 35482005 DOI: 10.1083/jcb.202108093] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 03/10/2022] [Accepted: 04/14/2022] [Indexed: 11/22/2022] Open
Abstract
Tissue-resident macrophages play essential functions in the maintenance of tissue homeostasis and repair. Recently, the endocardium has been reported as a de novo hemogenic site for the contribution of hematopoietic cells, including cardiac macrophages, during embryogenesis. These observations challenge the current consensus that hematopoiesis originates from the hemogenic endothelium within the yolk sac and dorsal aorta. Whether the developing endocardium has such a hemogenic potential requires further investigation. Here, we generated new genetic tools to trace endocardial cells and reassessed their potential contribution to hematopoietic cells in the developing heart. Fate-mapping analyses revealed that the endocardium contributed minimally to cardiac macrophages and circulating blood cells. Instead, cardiac macrophages were mainly derived from the endothelium during primitive/transient definitive (yolk sac) and definitive (dorsal aorta) hematopoiesis. Our findings refute the concept of endocardial hematopoiesis, suggesting that the developing endocardium gives rise minimally to hematopoietic cells, including cardiac macrophages.
Collapse
Affiliation(s)
- Kuo Liu
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.,State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hengwei Jin
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Muxue Tang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shaohua Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xueying Tian
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Mingjun Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ximeng Han
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiuxiu Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Juan Tang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenjuan Pu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Zhongzhou Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Cardiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School and Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Bin Zhou
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.,State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental & Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
10
|
Feulner L, van Vliet PP, Puceat M, Andelfinger G. Endocardial Regulation of Cardiac Development. J Cardiovasc Dev Dis 2022; 9:jcdd9050122. [PMID: 35621833 PMCID: PMC9144171 DOI: 10.3390/jcdd9050122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 01/16/2023] Open
Abstract
The endocardium is a specialized form of endothelium that lines the inner side of the heart chambers and plays a crucial role in cardiac development. While comparatively less studied than other cardiac cell types, much progress has been made in understanding the regulation of and by the endocardium over the past two decades. In this review, we will summarize what is currently known regarding endocardial origin and development, the relationship between endocardium and other cardiac cell types, and the various lineages that endocardial cells derive from and contribute to. These processes are driven by key molecular mechanisms such as Notch and BMP signaling. These pathways in particular have been well studied, but other signaling pathways and mechanical cues also play important roles. Finally, we will touch on the contribution of stem cell modeling in combination with single cell sequencing and its potential translational impact for congenital heart defects such as bicuspid aortic valves and hypoplastic left heart syndrome. The detailed understanding of cellular and molecular processes in the endocardium will be vital to further develop representative stem cell-derived models for disease modeling and regenerative medicine in the future.
Collapse
Affiliation(s)
- Lara Feulner
- Cardiovascular Genetics, CHU Sainte-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (L.F.); (P.P.v.V.)
- Department of Molecular Biology, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Patrick Piet van Vliet
- Cardiovascular Genetics, CHU Sainte-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (L.F.); (P.P.v.V.)
- LIA (International Associated Laboratory) CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada;
- LIA (International Associated Laboratory) INSERM, 13885 Marseille, France
| | - Michel Puceat
- LIA (International Associated Laboratory) CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada;
- LIA (International Associated Laboratory) INSERM, 13885 Marseille, France
- INSERM U-1251, Marseille Medical Genetics, Aix-Marseille University, 13885 Marseille, France
| | - Gregor Andelfinger
- Cardiovascular Genetics, CHU Sainte-Justine Research Centre, Montreal, QC H3T 1C5, Canada; (L.F.); (P.P.v.V.)
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC H3T 1J4, Canada
- Department of Biochemistry, University of Montreal, Montreal, QC H3T 1J4, Canada
- Correspondence:
| |
Collapse
|
11
|
Lu Z, Jiang Z, Tang J, Lin C, Zhang H. Functions and origins of cardiac fat. FEBS J 2022; 290:1705-1718. [PMID: 35114069 DOI: 10.1111/febs.16388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/06/2022] [Accepted: 02/02/2022] [Indexed: 11/28/2022]
Abstract
Triglyceride droplets can be stored within cardiac adipocytes (CAs) and cardiomyocytes in the heart. Cardiac adipocytes reside in three distinct regions: pericardial, epicardial, and intramyocardial adipose tissues. In healthy individuals, cardiac adipose tissues modulate cardiovascular functions and energy partitioning, which are, thus, protective. However, ectopic deposition of cardiac adipose tissues turns them into adverse lipotoxic, prothrombotic, and pro-inflammatory tissues with local and systemic contribution to the development of cardiovascular disorders. Accumulation of triglyceride droplets in cardiomyocytes may lead to lipotoxic injury of cardiomyocytes and contribute to the development of cardiac hypertrophy and dysfunction. Here, we summarize the roles of CAs and myocardial triglyceride droplets under physiological and pathological conditions and review the cellular sources of CAs in heart development and diseases. Understanding the functions and cellular origins of cardiac fat will provide clues for future studies on pathophysiological processes and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Zhengkai Lu
- School of Life Science and Technology ShanghaiTech University China
- University of Chinese Academy of Sciences Beijing China
| | - Zhen Jiang
- School of Life Science and Technology ShanghaiTech University China
| | - Juan Tang
- Institute for Regenerative Medicine Shanghai East Hospital Frontier Science Center for Stem Cell Research School of Life Science and Technology Tongji University Shanghai China
| | - Chao‐Po Lin
- School of Life Science and Technology ShanghaiTech University China
| | - Hui Zhang
- School of Life Science and Technology ShanghaiTech University China
| |
Collapse
|
12
|
Tang J, Zhu H, Tian X, Wang H, Liu S, Liu K, Zhao H, He L, Huang X, Feng Z, Ding Z, Long B, Yan Y, Smart N, Gong H, Luo Q, Zhou B. Extension of Endocardium-Derived Vessels Generate Coronary Arteries in Neonates. Circ Res 2022; 130:352-365. [PMID: 34995101 DOI: 10.1161/circresaha.121.320335] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Unraveling how new coronary arteries develop may provide critical information for establishing novel therapeutic approaches to treating ischemic cardiac diseases. There are two distinct coronary vascular populations derived from different origins in the developing heart. Understanding the formation of coronary arteries may provide insights into new ways of promoting coronary artery formation after myocardial infarction. Methods: To understand how intramyocardial coronary arteries are generated to connect these two coronary vascular populations, we combined genetic lineage tracing, light-sheet microscopy, fluorescence micro-optical sectioning tomography, and tissue-specific gene knockout approaches to understand their cellular and molecular mechanisms. Results: We show that a subset of intramyocardial coronary arteries form by angiogenic extension of endocardium-derived vascular tunnels in the neonatal heart. Three-dimensional whole-mount fluorescence imaging showed that these endocardium-derived vascular tunnels or tubes adopt an arterial fate in neonates. Mechanistically, we implicate Mettl3 and Notch signaling in regulating endocardium-derived intramyocardial coronary artery formation. Functionally, these intramyocardial arteries persist into adulthood and play a protective role after myocardial infarction. Conclusions: A subset of intramyocardial coronary arteries form by extension of endocardium-derived vascular tunnels in the neonatal heart.
Collapse
Affiliation(s)
- Juan Tang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Huan Zhu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Xueying Tian
- Key Laboratory of Regenerative Medicine of Ministry of Education, Department of Developmental and Regenerative Biology, College of Life Science and Technology, Jinan University, Guangzhou, China (X.T.)
| | - Haixiao Wang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Shaoyan Liu
- Zhongshan Hospital, Fudan University, Shanghai, China (S.L., Y.Y.)
| | - Kuo Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Huan Zhao
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Lingjuan He
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Xiuzhen Huang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
| | - Zhao Feng
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China (Z.F., H.G.)
| | - Zhangheng Ding
- Britton Chance Center for Biomedical Photonics, MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China (Z.D., H.G.)
| | - Ben Long
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China (B.L., Q.L.)
| | - Yan Yan
- Zhongshan Hospital, Fudan University, Shanghai, China (S.L., Y.Y.)
| | - Nicola Smart
- British Heart Foundation Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford (N.S.)
| | - Hui Gong
- HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China (Z.F., H.G.)
- Britton Chance Center for Biomedical Photonics, MoE Key Laboratory for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China (Z.D., H.G.)
| | - Qingming Luo
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China (B.L., Q.L.)
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences (J.T., H.Z., H.W., K.L., H.Z., L.H., X.H., B.Z.), University of Chinese Academy of Sciences, Beijing, China
- School of Life Science, Hangzhou Institute for Advanced Study (B.Z.), University of Chinese Academy of Sciences, Beijing, China
- School of Life Science and Technology, ShanghaiTech University, China (B.Z.)
| |
Collapse
|
13
|
Liu X, Liu L, Zhao J, Wang H, Li Y. Mechanotransduction regulates inflammation responses of epicardial adipocytes in cardiovascular diseases. Front Endocrinol (Lausanne) 2022; 13:1080383. [PMID: 36589802 PMCID: PMC9800500 DOI: 10.3389/fendo.2022.1080383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
Adipose tissue is a crucial regulator in maintaining cardiovascular homeostasis by secreting various bioactive products to mediate the physiological function of the cardiovascular system. Accumulating evidence shows that adipose tissue disorders contribute to several kinds of cardiovascular disease (CVD). Furthermore, the adipose tissue would present various biological effects depending on its tissue localization and metabolic statuses, deciding the individual cardiometabolic risk. Crosstalk between adipose and myocardial tissue is involved in the pathophysiological process of arrhythmogenic right ventricular cardiomyopathy (ARVC), cardiac fibrosis, heart failure, and myocardial infarction/atherosclerosis. The abnormal distribution of adipose tissue in the heart might yield direct and/or indirect effects on cardiac function. Moreover, mechanical transduction is critical for adipocytes in differentiation, proliferation, functional maturity, and homeostasis maintenance. Therefore, understanding the features of mechanotransduction pathways in the cellular ontogeny of adipose tissue is vital for underlining the development of adipocytes involved in cardiovascular disorders, which would preliminarily contribute positive implications on a novel therapeutic invention for cardiovascular diseases. In this review, we aim to clarify the role of mechanical stress in cardiac adipocyte homeostasis and its interplay with maintaining cardiac function.
Collapse
Affiliation(s)
- Xiaoliang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lei Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junfei Zhao
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Hua Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education (MOE), Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- *Correspondence: Yifei Li, ; Junfei Zhao, ; Hua Wang,
| |
Collapse
|
14
|
Zhang S, Li Y, Huang X, Liu K, Wang QD, Chen AF, Sun K, Lui KO, Zhou B. Seamless Genetic Recording of Transiently Activated Mesenchymal Gene Expression in Endothelial Cells During Cardiac Fibrosis. Circulation 2021; 144:2004-2020. [PMID: 34797683 DOI: 10.1161/circulationaha.121.055417] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: Cardiac fibrosis is a lethal outcome of excessive formation of myofibroblasts that are scar-forming cells accumulated after heart injury. It has been reported that cardiac endothelial cells (ECs) contribute to a substantial portion of myofibroblasts through EndoMT. Recent lineage tracing studies demonstrate that myofibroblasts are derived from expansion of resident fibroblasts rather than from transdifferentiation of ECs. However, it remains unknown whether ECs can transdifferentiate into myofibroblasts reversibly or EndoMT genes were just transiently activated in ECs during cardiac fibrosis. Methods: By using the dual recombination technology based on Cre-loxP and Dre-rox, we generated a genetic lineage tracing system for tracking EndoMT in cardiac ECs. We used it to examine if there is transiently activated mesenchymal gene expression in ECs during cardiac fibrosis. Activation of the broadly used marker gene in myofibroblasts, αSMA, and the transcription factor that induces epithelial to mesenchymal transition (EMT), Zeb1, was examined. Results: The genetic system enables continuous tracing of transcriptional activity of targeted genes in vivo. Our genetic fate mapping results revealed that a subset of cardiac ECs transiently expressed αSMA and Zeb1 during embryonic valve formation and transdifferentiated into mesenchymal cells through EndoMT. Nonetheless, they did not contribute to myofibroblasts; nor transiently expressed αSMA or Zeb1 after heart injury. Instead, expression of αSMA was activated in resident fibroblasts during cardiac fibrosis. Conclusions: Mesenchymal gene expression is activated in cardiac ECs through EndoMT in the developing heart; but ECs do not transdifferentiate into myofibroblasts, nor transiently express some known mesenchymal genes during homeostasis and fibrosis in the adult heart. Resident fibroblasts that are converted to myofibroblasts by activating mesenchymal gene expression are the major contributors to cardiac fibrosis.
Collapse
Affiliation(s)
- Shaohua Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiuzhen Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Kuo Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200092, China
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kathy O Lui
- Department of Chemical Pathology; and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai, 200031, China; School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| |
Collapse
|
15
|
Xie Y, Chen D, Jiang K, Song L, Qian N, Du Y, Yang Y, Wang F, Chen T. Hair shaft miniaturization causes stem cell depletion through mechanosensory signals mediated by a Piezo1-calcium-TNF-α axis. Cell Stem Cell 2021; 29:70-85.e6. [PMID: 34624205 DOI: 10.1016/j.stem.2021.09.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 07/19/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022]
Abstract
In aging, androgenic alopecia, and genetic hypotrichosis disorders, hair shaft miniaturization is often associated with hair follicle stem cell (HFSC) loss. However, the mechanism causing this stem cell depletion in vivo remains elusive. Here we show that hair shaft loss or a reduction in diameter shrinks the physical niche size, which results in mechanical compression of HFSCs and their apoptotic loss. Mechanistically, cell compression activates the mechanosensitive channel Piezo1, which triggers calcium influx. This confers tumor necrosis factor alpha (TNF-α) sensitivity in a hair-cycle-dependent manner in otherwise resistant HFSCs and induces ectopic apoptosis. Persistent hair shaft miniaturization during aging and genetic hypotrichosis disorders causes long-term HFSC loss by inducing continuous ectopic apoptosis through Piezo1. Our results identify an unconventional role of the inert hair shaft structure as a functional niche component governing HFSC survival and reveal a mechanosensory axis that regulates physical-niche-atrophy-induced stem cell depletion in vivo.
Collapse
Affiliation(s)
- Yuhua Xie
- China Agricultural University, Beijing, China; National Institute of Biological Sciences, Beijing, China
| | - Daoming Chen
- National Institute of Biological Sciences, Beijing, China
| | - Kaiju Jiang
- National Institute of Biological Sciences, Beijing, China
| | - Lifang Song
- National Institute of Biological Sciences, Beijing, China
| | - Nannan Qian
- National Institute of Biological Sciences, Beijing, China
| | - Yingxue Du
- National Institute of Biological Sciences, Beijing, China
| | - Yong Yang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, China
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China.
| |
Collapse
|
16
|
Cell proliferation fate mapping reveals regional cardiomyocyte cell-cycle activity in subendocardial muscle of left ventricle. Nat Commun 2021; 12:5784. [PMID: 34599161 PMCID: PMC8486850 DOI: 10.1038/s41467-021-25933-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 09/07/2021] [Indexed: 12/29/2022] Open
Abstract
Cardiac regeneration involves the generation of new cardiomyocytes from cycling cardiomyocytes. Understanding cell-cycle activity of pre-existing cardiomyocytes provides valuable information to heart repair and regeneration. However, the anatomical locations and in situ dynamics of cycling cardiomyocytes remain unclear. Here we develop a genetic approach for a temporally seamless recording of cardiomyocyte-specific cell-cycle activity in vivo. We find that the majority of cycling cardiomyocytes are positioned in the subendocardial muscle of the left ventricle, especially in the papillary muscles. Clonal analysis revealed that a subset of cycling cardiomyocytes have undergone cell division. Myocardial infarction and cardiac pressure overload induce regional patterns of cycling cardiomyocytes. Mechanistically, cardiomyocyte cell cycle activity requires the Hippo pathway effector YAP. These genetic fate-mapping studies advance our basic understanding of cardiomyocyte cell cycle activity and generation in cardiac homeostasis, repair, and regeneration. The adult mammalian heart exhibits stubbornly low levels of cardiomyocyte proliferation, leading to high morbidity after injury or heart attack. Here the authors develop an approach for tracking cardiomyocyte cell cycling and show that the majority are located adjacent to the endocardium.
Collapse
|
17
|
He L, Pu W, Liu X, Zhang Z, Han M, Li Y, Huang X, Han X, Li Y, Liu K, Shi M, Lai L, Sun R, Wang QD, Ji Y, Tchorz JS, Zhou B. Proliferation tracing reveals regional hepatocyte generation in liver homeostasis and repair. Science 2021; 371:371/6532/eabc4346. [PMID: 33632818 DOI: 10.1126/science.abc4346] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022]
Abstract
Organ homeostasis is orchestrated by time- and spatially restricted cell proliferation. Studies identifying cells with superior proliferative capacities often rely on the lineage tracing of a subset of cell populations, which introduces a potential selective bias. In this work, we developed a genetic system [proliferation tracer (ProTracer)] by incorporating dual recombinases to seamlessly record the proliferation events of entire cell populations over time in multiple organs. In the mouse liver, ProTracer revealed more hepatocyte proliferation in distinct zones during liver homeostasis, injury repair, and regrowth. Clonal analysis showed that most of the hepatocytes labeled by ProTracer had undergone cell division. By genetically recording proliferation events of entire cell populations, ProTracer enables the unbiased detection of specific cellular compartments with enhanced regenerative capacities.
Collapse
Affiliation(s)
- Lingjuan He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenjuan Pu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuxiu Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhenqian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Maoying Han
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yi Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuzhen Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ximeng Han
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kuo Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Mengyang Shi
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Liang Lai
- Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Ruilin Sun
- Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Qing-Dong Wang
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Yong Ji
- The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Jan S Tchorz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Cardiac cell type-specific responses to injury and contributions to heart regeneration. CELL REGENERATION 2021; 10:4. [PMID: 33527149 PMCID: PMC7851195 DOI: 10.1186/s13619-020-00065-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
Heart disease is the leading cause of mortality worldwide. Due to the limited proliferation rate of mature cardiomyocytes, adult mammalian hearts are unable to regenerate damaged cardiac muscle following injury. Instead, injured area is replaced by fibrotic scar tissue, which may lead to irreversible cardiac remodeling and organ failure. In contrast, adult zebrafish and neonatal mammalian possess the capacity for heart regeneration and have been widely used as experimental models. Recent studies have shown that multiple types of cells within the heart can respond to injury with the activation of distinct signaling pathways. Determining the specific contributions of each cell type is essential for our understanding of the regeneration network organization throughout the heart. In this review, we provide an overview of the distinct functions and coordinated cell behaviors of several major cell types including cardiomyocytes, endocardial cells, epicardial cells, fibroblasts, and immune cells. The topic focuses on their specific responses and cellular plasticity after injury, and potential therapeutic applications.
Collapse
|
19
|
Jiang Z, Feng T, Lu Z, Wei Y, Meng J, Lin CP, Zhou B, Liu C, Zhang H. PDGFRb + mesenchymal cells, but not NG2 + mural cells, contribute to cardiac fat. Cell Rep 2021; 34:108697. [PMID: 33535029 DOI: 10.1016/j.celrep.2021.108697] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/01/2020] [Accepted: 01/06/2021] [Indexed: 12/19/2022] Open
Abstract
Understanding cellular origins of cardiac adipocytes (CAs) can offer important implications for the treatment of fat-associated cardiovascular diseases. Here, we perform lineage tracing studies by using various genetic models and find that cardiac mesenchymal cells (MCs) contribute to CAs in postnatal development and adult homeostasis. Although PDGFRa+ and PDGFRb+ MCs both give rise to intramyocardial adipocytes, PDGFRb+ MCs are demonstrated to be the major source of intramyocardial adipocytes. Moreover, we find that PDGFRb+ cells are heterogenous, as PDGFRb is expressed not only in pericytes and smooth muscle cells (SMCs) but also in some subendocardial, pericapillary, or adventitial PDGFRa+ fibroblasts. Dual-recombinase-mediated intersectional genetic lineage tracing reveals that PDGFRa+PDGFRb+ double-positive periendothelial fibroblasts contribute to intramyocardial adipocytes. In contrast, SMCs and NG2+ pericytes do not contribute to CAs. These in vivo findings demonstrate that PDGFRb+ MCs, but not NG2+ coronary vascular mural cells, are the major source of intramyocardial adipocytes.
Collapse
Affiliation(s)
- Zhen Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Teng Feng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengkai Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanxin Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jufeng Meng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Bin Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chen Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
20
|
Tian X, He L, Liu K, Pu W, Zhao H, Li Y, Liu X, Tang M, Sun R, Fei J, Ji Y, Qiao Z, Lui KO, Zhou B. Generation of a self-cleaved inducible Cre recombinase for efficient temporal genetic manipulation. EMBO J 2020; 39:e102675. [PMID: 31943281 DOI: 10.15252/embj.2019102675] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 01/31/2023] Open
Abstract
Site-specific recombinase-mediated genetic technology, such as inducible Cre-loxP recombination (CreER), is widely used for in vivo genetic manipulation with temporal control. The Cre-loxP technology improves our understanding on the in vivo function of specific genes in organ development, tissue regeneration, and disease progression. However, inducible CreER often remains inefficient in gene deletion. In order to improve the efficiency of gene manipulation, we generated a self-cleaved inducible CreER (sCreER) that switches inducible CreER into a constitutively active Cre by itself. We generated endocardial driver Npr3-sCreER and fibroblast driver Col1a2-sCreER, and compared them with conventional Npr3-CreER and Col1a2-CreER, respectively. For easy-to-recombine alleles such as R26-tdTomato, there was no significant difference in recombination efficiency between sCreER and the conventional CreER. However, for alleles that were relatively inert for recombination such as R26-Confetti, R26-LZLT, R26-GFP, or VEGFR2flox/flox alleles, sCreER showed a significantly higher efficiency in recombination compared with conventional CreER in endocardial cells or fibroblasts. Compared with conventional CreER, sCreER significantly enhances the efficiency of recombination to induce gene expression or gene deletion, allowing temporal yet effective in vivo genomic modification for studying gene function in specific cell lineages.
Collapse
Affiliation(s)
- Xueying Tian
- Key Laboratory of Regenerative Medicine of Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China.,The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lingjuan He
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kuo Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Wenjuan Pu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huan Zhao
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuxiu Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Muxue Tang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ruilin Sun
- Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Jian Fei
- Shanghai Model Organisms Center, Inc., Shanghai, China
| | - Yong Ji
- The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China.,Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Zengyong Qiao
- Department of Cardiovascular Medicine, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Sha Tin, Hong Kong SAR, China
| | - Bin Zhou
- Key Laboratory of Regenerative Medicine of Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China.,The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China.,The Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Liu K, Tang M, Jin H, Liu Q, He L, Zhu H, Liu X, Han X, Li Y, Zhang L, Tang J, Pu W, Lv Z, Wang H, Ji H, Zhou B. Triple-cell lineage tracing by a dual reporter on a single allele. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49927-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
22
|
Liu K, Tang M, Jin H, Liu Q, He L, Zhu H, Liu X, Han X, Li Y, Zhang L, Tang J, Pu W, Lv Z, Wang H, Ji H, Zhou B. Triple-cell lineage tracing by a dual reporter on a single allele. J Biol Chem 2019; 295:690-700. [PMID: 31771978 DOI: 10.1074/jbc.ra119.011349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/01/2019] [Indexed: 12/12/2022] Open
Abstract
Genetic lineage tracing is widely used to study organ development and tissue regeneration. Multicolor reporters are a powerful platform for simultaneously tracking discrete cell populations. Here, combining Dre-rox and Cre-loxP systems, we generated a new dual-recombinase reporter system, called Rosa26 traffic light reporter (R26-TLR), to monitor red, green, and yellow fluorescence. Using this new reporter system with the three distinct fluorescent reporters combined on one allele, we found that the readouts of the two recombinases Cre and Dre simultaneously reflect Cre+Dre-, Cre-Dre+, and Cre+Dre+ cell lineages. As proof of principle, we show specific labeling in three distinct progenitor/stem cell populations, including club cells, AT2 cells, and bronchoalveolar stem cells, in Sftpc-DreER;Scgb1a1-CreER;R26-TLR mice. By using this new dual-recombinase reporter system, we simultaneously traced the cell fate of these three distinct cell populations during lung repair and regeneration, providing a more comprehensive picture of stem cell function in distal airway repair and regeneration. We propose that this new reporter system will advance developmental and regenerative research by facilitating a more sophisticated genetic approach to studying in vivo cell fate plasticity.
Collapse
Affiliation(s)
- Kuo Liu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Muxue Tang
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Hengwei Jin
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Qiaozhen Liu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Lingjuan He
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Huan Zhu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Xiuxiu Liu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Ximeng Han
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yan Li
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Libo Zhang
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Juan Tang
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Wenjuan Pu
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Zan Lv
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Haixiao Wang
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Hongbin Ji
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, University of the Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China .,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
23
|
Abstract
Endothelial cells and mesenchymal cells are two different cell types with distinct morphologies, phenotypes, functions, and gene profiles. Accumulating evidence, notably from lineage-tracing studies, indicates that the two cell types convert into each other during cardiovascular development and pathogenesis. During heart development, endothelial cells transdifferentiate into mesenchymal cells in the endocardial cushion through endothelial-to-mesenchymal transition (EndoMT), a process that is critical for the formation of cardiac valves. Studies have also reported that EndoMT contributes to the development of various cardiovascular diseases, including myocardial infarction, cardiac fibrosis, valve calcification, endocardial elastofibrosis, atherosclerosis, and pulmonary arterial hypertension. Conversely, cardiac fibroblasts can transdifferentiate into endothelial cells and contribute to neovascularization after cardiac injury. However, progress in genetic lineage tracing has challenged the role of EndoMT, or its reversed programme, in the development of cardiovascular diseases. In this Review, we discuss the caveats of using genetic lineage-tracing technology to investigate cell-lineage conversion; we also reassess the role of EndoMT in cardiovascular development and diseases and elaborate on the molecular signals that orchestrate EndoMT in pathophysiological processes. Understanding the role and mechanisms of EndoMT in diseases will unravel the therapeutic potential of targeting this process and will provide a new paradigm for the development of regenerative medicine to treat cardiovascular diseases.
Collapse
|
24
|
Abstract
Endocardial cells are specialized endothelial cells that form the innermost layer of the heart wall. By virtue of genetic lineage-tracing technology, many of the unexpected roles of endocardium during murine heart development, diseases, and regeneration have been identified recently. In addition to heart valves developed from the well-known endothelial to mesenchymal transition, recent fate-mapping studies using mouse models reveal that multiple cardiac cell lineages are also originated from the endocardium. This review focuses on a variety of different cell types that are recently reported to be endocardium derived during murine heart development, diseases, and regeneration. These multiple cell fates underpin the unprecedented roles of endocardial progenitors in function, pathological progression, and regeneration of the heart. Because emerging studies suggest that developmental mechanisms can be redeployed and recapitulated in promoting heart disease development and also cardiac repair and regeneration, understanding the mechanistic regulation of endocardial plasticity and modulation of their cell fate conversion may uncover new therapeutic potential in facilitating heart regeneration.
Collapse
Affiliation(s)
- Hui Zhang
- From the The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.Z., B.Z.); School of Life Science and Technology, ShanghaiTech University, China (H.Z., B.Z.); Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China (K.O.L.); and Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).
| | - Kathy O Lui
- From the The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.Z., B.Z.); School of Life Science and Technology, ShanghaiTech University, China (H.Z., B.Z.); Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China (K.O.L.); and Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).
| | - Bin Zhou
- From the The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, China (H.Z., B.Z.); School of Life Science and Technology, ShanghaiTech University, China (H.Z., B.Z.); Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, China (K.O.L.); and Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China (B.Z.).
| |
Collapse
|
25
|
Huang X, Feng T, Jiang Z, Meng J, Kou S, Lu Z, Chen W, Lin CP, Zhou B, Zhang H. Dual lineage tracing identifies intermediate mesenchymal stage for endocardial contribution to fibroblasts, coronary mural cells, and adipocytes. J Biol Chem 2019; 294:8894-8906. [PMID: 31010826 DOI: 10.1074/jbc.ra118.006994] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/04/2019] [Indexed: 11/06/2022] Open
Abstract
Early embryonic endocardium undergoes endothelial-to-mesenchymal transition to form cardiac cushion mesenchymal cells (MCs). Embryonic endocardium also gives rise to fibroblasts, intramyocardial adipocytes, and coronary mural cells, including smooth muscle cells and pericytes, in development. Whether endocardial cells directly differentiate into fibroblasts, coronary mural cells, and adipocytes or indirectly via an intermediate stage of endocardial-derived cushion MCs remains unknown. In addition to endocardium, epicardium and neural crest also contribute to cardiac cushion MCs. Given the developmental heterogeneity of cushion MCs and the lack of specific markers for endocardial-derived cushion MCs, conventional genetic lineage tracing utilizing Cre recombinase driven by one specific regulatory element is not sufficient to examine the fates of endocardial-derived cushion MCs. Intersectional genetic targeting approaches, which combine regulatory elements from two or more genes, have been employed to increase the specificity of cell targeting. Here, we developed a dual-recombinase intersectional targeting approach using Nfatc1-Dre, Sox9-CreER, and Cre/Dre double-dependent reporter Ai66 to specifically label endocardial-derived cushion MCs. Taking advantage of intersectional lineage tracing, we found that a subset of cardiac cells including fibroblasts, coronary mural cells, and intramyocardial adipocytes in adult hearts were derived from endocardial-derived cushion MCs. Our study suggests that embryonic endocardium contributes to cushion MCs first, and then endocardial-derived cushion MCs migrate into myocardium and differentiate into fibroblasts, coronary mural cells, and adipocytes in development. Understanding developmental origins of cardiac cell lineages will provide us more insights into cardiac development, regeneration, and diseases.
Collapse
Affiliation(s)
- Xinyan Huang
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Teng Feng
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Jiang
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jufeng Meng
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shan Kou
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhengkai Lu
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,the University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weize Chen
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chao-Po Lin
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Bin Zhou
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.,the University of Chinese Academy of Sciences, Beijing 100049, China.,the State Key Laboratory of Cell Biology, Chinese Academy of Sciences Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China, and.,the Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Hui Zhang
- From the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China,
| |
Collapse
|
26
|
Liu Q, Liu K, Cui G, Huang X, Yao S, Guo W, Qin Z, Li Y, Yang R, Pu W, Zhang L, He L, Zhao H, Yu W, Tang M, Tian X, Cai D, Nie Y, Hu S, Ren T, Qiao Z, Huang H, Zeng YA, Jing N, Peng G, Ji H, Zhou B. Lung regeneration by multipotent stem cells residing at the bronchioalveolar-duct junction. Nat Genet 2019; 51:728-738. [DOI: 10.1038/s41588-019-0346-6] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023]
|
27
|
Kovacic JC, Dimmeler S, Harvey RP, Finkel T, Aikawa E, Krenning G, Baker AH. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J Am Coll Cardiol 2019; 73:190-209. [PMID: 30654892 PMCID: PMC6865825 DOI: 10.1016/j.jacc.2018.09.089] [Citation(s) in RCA: 405] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/20/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022]
Abstract
Endothelial to mesenchymal transition (EndMT) is a process whereby an endothelial cell undergoes a series of molecular events that lead to a change in phenotype toward a mesenchymal cell (e.g., myofibroblast, smooth muscle cell). EndMT plays a fundamental role during development, and mounting evidence indicates that EndMT is involved in adult cardiovascular diseases (CVDs), including atherosclerosis, pulmonary hypertension, valvular disease, and fibroelastosis. Therefore, the targeting of EndMT may hold therapeutic promise for treating CVD. However, the field faces a number of challenges, including the lack of a precise functional and molecular definition, a lack of understanding of the causative pathological role of EndMT in CVDs (versus being a "bystander-phenomenon"), and a lack of robust human data corroborating the extent and causality of EndMT in adult CVDs. Here, we review this emerging but exciting field, and propose a framework for its systematic advancement at the molecular and translational levels.
Collapse
Affiliation(s)
- Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, and German Center of Cardiovascular Research, Frankfurt, Germany
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia; St. Vincent's Clinical School and School of Biotechnology and Biomolecular Science, University of New South Wales, Kensington, New South Wales, Australia
| | - Toren Finkel
- Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, and Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Andrew H Baker
- UoE/BHF Center for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, United Kingdom.
| |
Collapse
|
28
|
Liu K, Yu W, Tang M, Tang J, Liu X, Liu Q, Li Y, He L, Zhang L, Evans SM, Tian X, Lui KO, Zhou B. A dual genetic tracing system identifies diverse and dynamic origins of cardiac valve mesenchyme. Development 2018; 145:dev.167775. [PMID: 30111655 DOI: 10.1242/dev.167775] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/27/2018] [Indexed: 12/24/2022]
Abstract
In vivo genomic engineering is instrumental for studying developmental biology and regenerative medicine. Development of novel systems with more site-specific recombinases (SSRs) that complement with the commonly used Cre-loxP would be valuable for more precise lineage tracing and genome editing. Here, we introduce a new SSR system via Nigri-nox. By generating tissue-specific Nigri knock-in and its responding nox reporter mice, we show that the Nigri-nox system works efficiently in vivo by targeting specific tissues. As a new orthogonal system to Cre-loxP, Nigri-nox provides an additional control of genetic manipulation. We also demonstrate how the two orthogonal systems Nigri-nox and Cre-loxP could be used simultaneously to map the cell fate of two distinct developmental origins of cardiac valve mesenchyme in the mouse heart, providing dynamics of cellular contribution from different origins for cardiac valve mesenchyme during development. This work provides a proof-of-principle application of the Nigri-nox system for in vivo mouse genomic engineering. Coupled with other SSR systems, Nigri-nox would be valuable for more precise delineation of origins and cell fates during development, diseases and regeneration.
Collapse
Affiliation(s)
- Kuo Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Yu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Muxue Tang
- School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Juan Tang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiuxiu Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaozhen Liu
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Lingjuan He
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Libo Zhang
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Xueying Tian
- Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China
| | - Kathy O Lui
- Department of Chemical Pathology; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China .,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Key Laboratory of Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
29
|
Pu W, He L, Han X, Tian X, Li Y, Zhang H, Liu Q, Huang X, Zhang L, Wang QD, Yu Z, Yang X, Smart N, Zhou B. Genetic Targeting of Organ-Specific Blood Vessels. Circ Res 2018; 123:86-99. [PMID: 29764841 DOI: 10.1161/circresaha.118.312981] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/20/2023]
Abstract
RATIONALE Organs of the body require vascular networks to supply oxygen and nutrients and maintain physiological function. The blood vessels of different organs are structurally and functionally heterogeneous in nature. To more precisely dissect their distinct in vivo function in individual organs, without potential interference from off-site targets, it is necessary to genetically target them in an organ-specific manner. OBJECTIVE The objective of this study was to generate a genetic system that targets vascular endothelial cells in an organ- or tissue-specific manner and to exemplify the potential application of intersectional genetics for precise, target-specific gene manipulation in vivo. METHODS AND RESULTS We took advantage of 2 orthogonal recombination systems, Dre-rox and Cre-loxP, to create a genetic targeting system based on intersectional genetics. Using this approach, Cre activity was only detectable in cells that had expressed both Dre and Cre. Applying this new system, we generated a coronary endothelial cell-specific Cre (CoEC-Cre) and a brain endothelial cell-specific Cre (BEC-Cre). Through lineage tracing, gene knockout and overexpression experiments, we demonstrated that CoEC-Cre and BEC-Cre efficiently and specifically target blood vessels in the heart and brain, respectively. By deletion of vascular endothelial growth factor receptor 2 using BEC-Cre, we showed that vascular endothelial growth factor signaling regulates angiogenesis in the central nervous system and also controls the integrity of the blood-brain barrier. CONCLUSIONS We provide 2 examples to illustrate the use of intersectional genetics for more precise gene targeting in vivo, namely manipulation of genes in blood vessels of the heart and brain. More broadly, this system provides a valuable strategy for tissue-specific gene manipulation that can be widely applied to other fields of biomedical research.
Collapse
Affiliation(s)
- Wenjuan Pu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Lingjuan He
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Ximeng Han
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences; School of Life Science and Technology, Shanghai Tech University, China (X. Han, H.Z., B.Z.)
| | - Xueying Tian
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Yan Li
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Hui Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences; School of Life Science and Technology, Shanghai Tech University, China (X. Han, H.Z., B.Z.)
| | - Qiaozhen Liu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Xiuzhen Huang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Libo Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.).,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.)
| | - Qing-Dong Wang
- Bioscience Heart Failure, Cardiovascular and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden (Q.-D.W.)
| | - Zhenyang Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Laboratory of Proteomics, Institute of Biotechnology, China (Z.Y., X.Y.)
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Laboratory of Proteomics, Institute of Biotechnology, China (Z.Y., X.Y.)
| | - Nicola Smart
- British Heart Foundation Centre of Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom (N.S.)
| | - Bin Zhou
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences (W.P., L.H., X.T., Y.L., H.Z., Q.L., X. Huang, L.Z., B.Z.) .,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (W.P., L.H., X.T., Y.L., Q.L., X. Huang, L.Z., B.Z.).,Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences; School of Life Science and Technology, Shanghai Tech University, China (X. Han, H.Z., B.Z.).,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China (B.Z.)
| |
Collapse
|
30
|
Tang J, Zhang H, He L, Huang X, Li Y, Pu W, Yu W, Zhang L, Cai D, Lui KO, Zhou B. Genetic Fate Mapping Defines the Vascular Potential of Endocardial Cells in the Adult Heart. Circ Res 2018; 122:984-993. [DOI: 10.1161/circresaha.117.312354] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 01/22/2018] [Accepted: 01/25/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Juan Tang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Hui Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Lingjuan He
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Xiuzhen Huang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Yan Li
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Wenjuan Pu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Wei Yu
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Libo Zhang
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Dongqing Cai
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Kathy O. Lui
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| | - Bin Zhou
- From the State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.) and Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences (J.T., H.Z., L.H., X.H., Y.L., W.P., W.Y., L.Z., B.Z.), Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China; School of Life Science and Technology,
| |
Collapse
|
31
|
Enhancing the precision of genetic lineage tracing using dual recombinases. Nat Med 2017; 23:1488-1498. [PMID: 29131159 DOI: 10.1038/nm.4437] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 10/11/2017] [Indexed: 12/11/2022]
Abstract
The Cre-loxP recombination system is the most widely used technology for in vivo tracing of stem or progenitor cell lineages. The precision of this genetic system largely depends on the specificity of Cre recombinase expression in targeted stem or progenitor cells. However, Cre expression in nontargeted cell types can complicate the interpretation of lineage-tracing studies and has caused controversy in many previous studies. Here we describe a new genetic lineage tracing system that incorporates the Dre-rox recombination system to enhance the precision of conventional Cre-loxP-mediated lineage tracing. The Dre-rox system permits rigorous control of Cre-loxP recombination in lineage tracing, effectively circumventing potential uncertainty of the cell-type specificity of Cre expression. Using this new system we investigated two topics of recent debates-the contribution of c-Kit+ cardiac stem cells to cardiomyocytes in the heart and the contribution of Sox9+ hepatic progenitor cells to hepatocytes in the liver. By overcoming the technical hurdle of nonspecific Cre-loxP-mediated recombination, this new technology provides more precise analysis of cell lineage and fate decisions and facilitates the in vivo study of stem and progenitor cell plasticity in disease and regeneration.
Collapse
|
32
|
Fibroblasts in an endocardial fibroelastosis disease model mainly originate from mesenchymal derivatives of epicardium. Cell Res 2017; 27:1157-1177. [PMID: 28809397 DOI: 10.1038/cr.2017.103] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/18/2017] [Accepted: 06/29/2017] [Indexed: 02/06/2023] Open
Abstract
Endocardial fibroelastosis (EFE) refers to the thickening of the ventricular endocardium as a result of de novo deposition of subendocardial fibrous tissue layers during neonatal heart development. The origin of EFE fibroblasts is proposed to be postnatal endocardial cells that undergo an aberrant endothelial-to-mesenchymal transition (EndMT). Genetic lineage tracing of endocardial cells with the inducible endocardial Cre line Npr3-CreER and the endothelial cell tracing line Cdh5-CreER on an EFE-like model did not reveal any contribution of neonatal endocardial cells to fibroblasts in the EFE-like tissues. Instead, lineage tracing of embryonic epicardium by Wt1-CreER suggested that epicardium-derived mesenchymal cells (MCs) served as the major source of EFE fibroblasts. By labeling MCs using Sox9-CreER, we confirmed that MCs of the embryonic heart expand and contribute to the majority of neonatal EFE fibroblasts. During this pathological process, TGFβ signaling, the key mediator of fibroblasts activation, was highly upregulated in the EFE-like tissues. Targeting TGFβ signaling by administration of its antagonist bone morphogenetic protein 7 effectively reduced fibroblast accumulation and tissue fibrosis in the EFE-like model. Our study provides genetic evidence that excessive fibroblasts in the EFE-like tissues mainly originate from the epicardium-derived MCs through epicardial to mesenchymal transition (EpiMT). These EpiMT-derived fibroblasts within the EFE-like tissues could serve as a potential therapeutic target.
Collapse
|
33
|
He L, Huang X, Kanisicak O, Li Y, Wang Y, Li Y, Pu W, Liu Q, Zhang H, Tian X, Zhao H, Liu X, Zhang S, Nie Y, Hu S, Miao X, Wang QD, Wang F, Chen T, Xu Q, Lui KO, Molkentin JD, Zhou B. Preexisting endothelial cells mediate cardiac neovascularization after injury. J Clin Invest 2017. [PMID: 28650345 DOI: 10.1172/jci93868] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The mechanisms that promote the generation of new coronary vasculature during cardiac homeostasis and after injury remain a fundamental and clinically important area of study in the cardiovascular field. Recently, it was reported that mesenchymal-to-endothelial transition (MEndoT) contributes to substantial numbers of coronary endothelial cells after myocardial infarction. Therefore, the MEndoT has been proposed as a paradigm mediating neovascularization and is considered a promising therapeutic target in cardiac regeneration. Here, we show that preexisting endothelial cells mainly beget new coronary vessels in the adult mouse heart, with essentially no contribution from other cell sources through cell-lineage transdifferentiation. Genetic-lineage tracing revealed that cardiac fibroblasts expand substantially after injury, but do not contribute to the formation of new coronary blood vessels, indicating no contribution of MEndoT to neovascularization. Moreover, genetic-lineage tracing with a pulse-chase labeling strategy also showed that essentially all new coronary vessels in the injured heart are derived from preexisting endothelial cells, but not from other cell lineages. These data indicate that therapeutic strategies for inducing neovascularization should not be based on targeting presumptive lineage transdifferentiation such as MEndoT. Instead, preexisting endothelial cells appear more likely to be the therapeutic target for promoting neovascularization and driving heart regeneration after injury.
Collapse
Affiliation(s)
- Lingjuan He
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuzhen Huang
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Onur Kanisicak
- Department of Pediatrics and Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Yi Li
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Yue Wang
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Wenjuan Pu
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Qiaozhen Liu
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Hui Zhang
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Xueying Tian
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Huan Zhao
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Xiuxiu Liu
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Shaohua Zhang
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China
| | - Yu Nie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiang Miao
- Flow Cytometry Core Facility, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, CAS, Shanghai, China
| | - Qing-Dong Wang
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Clinical Development Biotech Unit, AstraZeneca, Mölndal, Sweden
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, China
| | - Ting Chen
- National Institute of Biological Sciences, Beijing, China
| | - Qingbo Xu
- Cardiovascular Division, British Heart Foundation Centre, King's College London, London, United Kingdom
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| | - Jeffery D Molkentin
- Department of Pediatrics and Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, CAS, University of Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
34
|
Gaborit B, Sengenes C, Ancel P, Jacquier A, Dutour A. Role of Epicardial Adipose Tissue in Health and Disease: A Matter of Fat? Compr Physiol 2017. [PMID: 28640452 DOI: 10.1002/cphy.c160034] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Epicardial adipose tissue (EAT) is a small but very biologically active ectopic fat depot that surrounds the heart. Given its rapid metabolism, thermogenic capacity, unique transcriptome, secretory profile, and simply measurability, epicardial fat has drawn increasing attention among researchers attempting to elucidate its putative role in health and cardiovascular diseases. The cellular crosstalk between epicardial adipocytes and cells of the vascular wall or myocytes is high and suggests a local role for this tissue. The balance between protective and proinflammatory/profibrotic cytokines, chemokines, and adipokines released by EAT seem to be a key element in atherogenesis and could represent a future therapeutic target. EAT amount has been found to predict clinical coronary outcomes. EAT can also modulate cardiac structure and function. Its amount has been associated with atrial fibrillation, coronary artery disease, and sleep apnea syndrome. Conversely, a beiging fat profile of EAT has been identified. In this review, we describe the current state of knowledge regarding the anatomy, physiology and pathophysiological role of EAT, and the factors more globally leading to ectopic fat development. We will also highlight the most recent findings on the origin of this ectopic tissue, and its association with cardiac diseases. © 2017 American Physiological Society. Compr Physiol 7:1051-1082, 2017.
Collapse
Affiliation(s)
- Bénédicte Gaborit
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| | - Coralie Sengenes
- STROMALab, Université de Toulouse, EFS, ENVT, Inserm U1031, ERL CNRS 5311, CHU Rangueil, Toulouse, France
| | - Patricia Ancel
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France
| | - Alexis Jacquier
- CNRS UMR 7339, Centre de Résonance Magnétique Biologique et Médicale (CRMBM), Marseille, France.,Radiology department, CHU La Timone, Marseille, France
| | - Anne Dutour
- NORT, Aix Marseille Univ, INSERM, INRA, NORT, Marseille, France.,Endocrinology Metabolic Diseases, and Nutrition Department, Pole ENDO, APHM, Aix-Marseille Univ, Marseille, France
| |
Collapse
|
35
|
Dueñas A, Aranega AE, Franco D. More than Just a Simple Cardiac Envelope; Cellular Contributions of the Epicardium. Front Cell Dev Biol 2017; 5:44. [PMID: 28507986 PMCID: PMC5410615 DOI: 10.3389/fcell.2017.00044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022] Open
Abstract
The adult pumping heart is formed by distinct tissue layers. From inside to outside, the heart is composed by an internal endothelial layer, dubbed the endocardium, a thick myocardial component which supports the pumping capacity of the heart and exteriorly covered by a thin mesothelial layer named the epicardium. Cardiac insults such as coronary artery obstruction lead to ischemia and thus to an irreversible damage of the myocardial layer, provoking in many cases heart failure and death. Thus, searching for new pathways to regenerate the myocardium is an urgent biomedical need. Interestingly, the capacity of heart regeneration is present in other species, ranging from fishes to neonatal mammals. In this context, several lines of evidences demonstrated a key regulatory role for the epicardial layer. In this manuscript, we provide a state-of-the-art review on the developmental process leading to the formation of the epicardium, the distinct pathways controlling epicardial precursor cell specification and determination and current evidences on the regenerative potential of the epicardium to heal the injured heart.
Collapse
Affiliation(s)
- Angel Dueñas
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| | - Amelia E Aranega
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| | - Diego Franco
- Cardiac and Skeletal Muscle Research Group, Department of Experimental Biology, University of JaénJaén, Spain
| |
Collapse
|
36
|
Wang Y, Huang X, He L, Pu W, Li Y, Liu Q, Li Y, Zhang L, Yu W, Zhao H, Zhou Y, Zhou B. Genetic tracing of hepatocytes in liver homeostasis, injury, and regeneration. J Biol Chem 2017; 292:8594-8604. [PMID: 28377509 DOI: 10.1074/jbc.m117.782029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/27/2017] [Indexed: 12/20/2022] Open
Abstract
The liver possesses a remarkable capacity to regenerate after damage. There is a heated debate on the origin of new hepatocytes after injuries in adult liver. Hepatic stem/progenitor cells have been proposed to produce functional hepatocytes after injury. Recent studies have argued against this model and suggested that pre-existing hepatocytes, rather than stem cells, contribute new hepatocytes. This hepatocyte-to-hepatocyte model is mainly based on labeling of hepatocytes with Cre-recombinase delivered by the adeno-associated virus. However, the impact of virus infection on cell fate determination, consistency of infection efficiency, and duration of Cre-virus in hepatocytes remain confounding factors that interfere with the data interpretation. Here, we generated a new genetic tool Alb-DreER to label almost all hepatocytes (>99.5%) and track their contribution to different cell lineages in the liver. By "pulse-and-chase" strategy, we found that pre-existing hepatocytes labeled by Alb-DreER contribute to almost all hepatocytes during normal homeostasis and after liver injury. Virtually all hepatocytes in the injured liver are descendants of pre-existing hepatocytes through self-expansion. We concluded that stem cell differentiation is unlikely to be responsible for the generation of a substantial number of new hepatocytes in adult liver. Our study also provides a new mouse tool for more precise in vivo genetic study of hepatocytes in the field.
Collapse
Affiliation(s)
- Yue Wang
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - XiuZhen Huang
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Lingjuan He
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenjuan Pu
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Li
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiaozhen Liu
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yi Li
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Libo Zhang
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Yu
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Huan Zhao
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingqun Zhou
- the Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China,
| | - Bin Zhou
- From The State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China, .,the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China.,the Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China, and.,the School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
37
|
Atrial natriuretic peptide regulates adipose tissue accumulation in adult atria. Proc Natl Acad Sci U S A 2017; 114:E771-E780. [PMID: 28096344 DOI: 10.1073/pnas.1610968114] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The abundance of epicardial adipose tissue (EAT) is associated with atrial fibrillation (AF), the most frequent cardiac arrhythmia. However, both the origin and the factors involved in EAT expansion are unknown. Here, we found that adult human atrial epicardial cells were highly adipogenic through an epithelial-mesenchymal transition both in vitro and in vivo. In a genetic lineage tracing the WT1CreERT2+/-RosatdT+/- mouse model subjected to a high-fat diet, adipocytes of atrial EAT derived from a subset of epicardial progenitors. Atrial myocardium secretome induces the adipogenic differentiation of adult mesenchymal epicardium-derived cells by modulating the balance between mesenchymal Wingless-type Mouse Mammary Tumor Virus integration site family, member 10B (Wnt10b)/β-catenin and adipogenic ERK/MAPK signaling pathways. The adipogenic property of the atrial secretome was enhanced in AF patients. The atrial natriuretic peptide secreted by atrial myocytes is a major adipogenic factor operating at a low concentration by binding to its natriuretic peptide receptor A (NPRA) receptor and, in turn, by activating a cGMP-dependent pathway. Hence, our data indicate cross-talk between EAT expansion and mechanical function of the atrial myocardium.
Collapse
|
38
|
Aldiss P, Davies G, Woods R, Budge H, Sacks HS, Symonds ME. 'Browning' the cardiac and peri-vascular adipose tissues to modulate cardiovascular risk. Int J Cardiol 2016; 228:265-274. [PMID: 27865196 PMCID: PMC5236060 DOI: 10.1016/j.ijcard.2016.11.074] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/05/2016] [Indexed: 01/02/2023]
Abstract
Excess visceral adiposity, in particular that located adjacent to the heart and coronary arteries is associated with increased cardiovascular risk. In the pathophysiological state, dysfunctional adipose tissue secretes an array of factors modulating vascular function and driving atherogenesis. Conversely, brown and beige adipose tissues utilise glucose and lipids to generate heat and are associated with improved cardiometabolic health. The cardiac and thoracic perivascular adipose tissues are now understood to be composed of brown adipose tissue in the healthy state and undergo a brown-to-white transition i.e. during obesity which may be a driving factor of cardiovascular disease. In this review we discuss the risks of excess cardiac and vascular adiposity and potential mechanisms by which restoring the brown phenotype i.e. “re-browning” could potentially be achieved in clinically relevant populations. Epicardial, paracardial and thoracic perivascular adipose tissues resemble BAT at birth. Despite ‘whitening’ in early life these depots remain metabolically active and potentially thermogenic into adulthood. Obesity induces further ‘whitening’ and inflammation in these depots likely driving the atherogenesis. Maintaining or inducing the brown phenotype in these depots could prevent atherosclerotic disease.
Collapse
Affiliation(s)
- Peter Aldiss
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Graeme Davies
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Rachel Woods
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Helen Budge
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH
| | - Harold S Sacks
- VA Greater Los Angeles Healthcare System, Endocrinology and Diabetes Division, and Department of Medicine David Geffen School of Medicine, Los Angeles, CA 90073, USA
| | - Michael E Symonds
- The Early Life Research Unit, Division of Child Health, Obstetrics and Gynaecology, School of Medicine, University Hospital, University of Nottingham, Nottingham, UK, NG7 2UH.
| |
Collapse
|
39
|
Liu L, Lei I, Karatas H, Li Y, Wang L, Gnatovskiy L, Dou Y, Wang S, Qian L, Wang Z. Targeting Mll1 H3K4 methyltransferase activity to guide cardiac lineage specific reprogramming of fibroblasts. Cell Discov 2016; 2:16036. [PMID: 27924221 PMCID: PMC5113048 DOI: 10.1038/celldisc.2016.36] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022] Open
Abstract
Generation of induced cardiomyocytes (iCMs) directly from fibroblasts offers a great opportunity for cardiac disease modeling and cardiac regeneration. A major challenge of iCM generation is the low conversion rate. To address this issue, we attempted to identify small molecules that could potentiate the reprogramming ability towards cardiac fate by removing inhibitory roadblocks. Using mouse embryonic fibroblasts as the starting cell source, we first screened 47 cardiac development related epigenetic and transcription factors, and identified an unexpected role of H3K4 methyltransferase Mll1 and related factor Men1 in inhibiting iCM reprogramming. We then applied small molecules (MM408 and MI503) of Mll1 pathway inhibitors and observed an improved efficiency in converting embryonic fibroblasts and cardiac fibroblasts into functional cardiomyocyte-like cells. We further observed that these inhibitors directly suppressed the expression of Mll1 target gene Ebf1 involved in adipocyte differentiation. Consequently, Mll1 inhibition significantly decreased the formation of adipocytes during iCM induction. Therefore, Mll1 inhibitors likely increased iCM efficiency by suppressing alternative lineage gene expression. Our studies show that targeting Mll1 dependent H3K4 methyltransferase activity provides specificity in the process of cardiac reprogramming. These findings shed new light on the molecular mechanisms underlying cardiac conversion of fibroblasts and provide novel targets and small molecules to improve iCM reprogramming for clinical applications.
Collapse
Affiliation(s)
- Liu Liu
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan, Ann Arbor, MI, USA; Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Hacer Karatas
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Yangbing Li
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Li Wang
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; McAllister Heart Institute University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Leonid Gnatovskiy
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| | - Yali Dou
- Department of Pathology, University of Michigan Medical School, 1301 Catherine , Ann Arbor, MI, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA; Department of Medicinal Chemistry, University of Michigan College of Pharmacy, Ann Arbor, MI, USA
| | - Li Qian
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA; McAllister Heart Institute University of North Carolina, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Frankel Cardiovascular Center, The University of Michigan , Ann Arbor, MI, USA
| |
Collapse
|
40
|
Vascular Development and Regeneration in the Mammalian Heart. J Cardiovasc Dev Dis 2016; 3:jcdd3020023. [PMID: 29367569 PMCID: PMC5715682 DOI: 10.3390/jcdd3020023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/31/2016] [Accepted: 06/14/2016] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases including coronary artery disease are the leading cause of death worldwide. Unraveling the developmental origin of coronary vessels could offer important therapeutic implications for treatment of cardiovascular diseases. The recent identification of the endocardial source of coronary vessels reveals a heterogeneous origin of coronary arteries in the adult heart. In this review, we will highlight recent advances in finding the sources of coronary vessels in the mammalian heart from lineage-tracing models as well as differentiation studies using pluripotent stem cells. Moreover, we will also discuss how we induce neovascularization in the damaged heart through transient yet highly efficient expression of VEGF-modified mRNAs as a potentially therapeutic delivery platform.
Collapse
|
41
|
Kawanishi K. Diverse properties of the mesothelial cells in health and disease. Pleura Peritoneum 2016; 1:79-89. [PMID: 30911611 DOI: 10.1515/pp-2016-0009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 05/19/2016] [Indexed: 12/17/2022] Open
Abstract
Mesothelial cells (MCs) form the superficial anatomic layer of serosal membranes, including pleura, pericardium, peritoneum, and the tunica of the reproductive organs. MCs produce a protective, non-adhesive barrier against physical and biochemical damages. MCs express a wide range of phenotypic markers, including vimentin and cytokeratins. MCs play key roles in fluid transport and inflammation, as reflected by the modulation of biochemical markers such as transporters, adhesion molecules, cytokines, growth factors, reactive oxygen species and their scavengers. MCs synthesize extracellular matrix related molecules, and the surface of MC microvilli secretes a highly hydrophilic protective barrier, "glycocalyx", consisting mainly of glycosaminoglycans. MCs maintain a balance between procoagulant and fibrinolytic activation by producing a whole range of regulators, can synthetize fibrin and therefore form adhesions. Synthesis and recognition of hyaluronan and sialic acids might be a new insight to explain immunoactive and immunoregulatory properties of MCs. Epithelial to mesenchymal transition of MCs may involve serosal repair and remodeling. MCs might also play a role in the development and remodeling of visceral adipose tissue. Taken together, MCs play important roles in health and disease in serosal cavities of the body. The mesothelium is not just a membrane and should be considered as an organ.
Collapse
|
42
|
Matyas C, Varga ZV, Mukhopadhyay P, Paloczi J, Lajtos T, Erdelyi K, Nemeth BT, Nan M, Hasko G, Gao B, Pacher P. Chronic plus binge ethanol feeding induces myocardial oxidative stress, mitochondrial and cardiovascular dysfunction, and steatosis. Am J Physiol Heart Circ Physiol 2016; 310:H1658-H1670. [PMID: 27106042 PMCID: PMC4935511 DOI: 10.1152/ajpheart.00214.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 04/15/2016] [Indexed: 12/31/2022]
Abstract
Alcoholic cardiomyopathy in humans develops in response to chronic excessive alcohol consumption; however, good models of alcohol-induced cardiomyopathy in mice are lacking. Herein we describe mouse models of alcoholic cardiomyopathies induced by chronic and binge ethanol (EtOH) feeding and characterize detailed hemodynamic alterations, mitochondrial function, and redox signaling in these models. Mice were fed a liquid diet containing 5% EtOH for 10, 20, and 40 days (d) combined with single or multiple EtOH binges (5 g/kg body wt). Isocalorically pair-fed mice served as controls. Left ventricular (LV) function and morphology were assessed by invasive pressure-volume conductance approach and by echocardiography. Mitochondrial complex (I, II, IV) activities, 3-nitrotyrosine (3-NT) levels, gene expression of markers of oxidative stress (gp91phox, p47phox), mitochondrial biogenesis (PGC1α, peroxisome proliferator-activated receptor α), and fibrosis were examined. Cardiac steatosis and fibrosis were investigated by histological/immunohistochemical methods. Chronic and binge EtOH feeding (already in 10 days EtOH plus single binge group) was characterized by contractile dysfunction (decreased slope of end-systolic pressure-volume relationship and preload recruitable stroke work), impaired relaxation (decreased time constant of LV pressure decay and maximal slope of systolic pressure decrement), and vascular dysfunction (impaired arterial elastance and lower total peripheral resistance). This was accompanied by enhanced myocardial oxidative/nitrative stress (3-NT; gp91phox; p47phox; angiotensin II receptor, type 1a) and deterioration of mitochondrial complex I, II, IV activities and mitochondrial biogenesis, excessive cardiac steatosis, and higher mortality. Collectively, chronic plus binge EtOH feeding in mice leads to alcohol-induced cardiomyopathies (National Institute on Alcohol Abuse and Alcoholism models) characterized by increased myocardial oxidative/nitrative stress, impaired mitochondrial function and biogenesis, and enhanced cardiac steatosis.
Collapse
Affiliation(s)
- Csaba Matyas
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland; Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Zoltan V Varga
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Partha Mukhopadhyay
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Janos Paloczi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Tamas Lajtos
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Katalin Erdelyi
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Balazs T Nemeth
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Mintong Nan
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Gyorgy Hasko
- Department of Surgery, Rutgers New Jersey Medical School, University Heights, Newark, New Jersey; and
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland;
| |
Collapse
|
43
|
Gardner JD. Alcohol binge drinking: getting to the heart of it. Am J Physiol Heart Circ Physiol 2016; 310:H1606-7. [PMID: 27208162 DOI: 10.1152/ajpheart.00368.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 01/02/2023]
Affiliation(s)
- J D Gardner
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
44
|
Zhang H, Pu W, Li G, Huang X, He L, Tian X, Liu Q, Zhang L, Wu SM, Sucov HM, Zhou B. Endocardium Minimally Contributes to Coronary Endothelium in the Embryonic Ventricular Free Walls. Circ Res 2016; 118:1880-93. [PMID: 27056912 DOI: 10.1161/circresaha.116.308749] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/07/2016] [Indexed: 12/24/2022]
Abstract
RATIONALE There is persistent uncertainty regarding the developmental origins of coronary vessels, with 2 principal sources suggested as ventricular endocardium or sinus venosus (SV). These 2 proposed origins implicate fundamentally distinct mechanisms of vessel formation. Resolution of this controversy is critical for deciphering the programs that result in the formation of coronary vessels and has implications for research on therapeutic angiogenesis. OBJECTIVE To resolve the controversy over the developmental origin of coronary vessels. METHODS AND RESULTS We first generated nuclear factor of activated T cells (Nfatc1)-Cre and Nfatc1-Dre lineage tracers for endocardium labeling. We found that Nfatc1 recombinases also label a significant portion of SV endothelial cells in addition to endocardium. Therefore, restricted endocardial lineage tracing requires a specific marker that distinguishes endocardium from SV. By single-cell gene expression analysis, we identified a novel endocardial gene natriuretic peptide receptor 3 (Npr3). Npr3 is expressed in the entirety of the endocardium but not in the SV. Genetic lineage tracing based on Npr3-CreER showed that endocardium contributes to a minority of coronary vessels in the free walls of embryonic heart. Intersectional genetic lineage tracing experiments demonstrated that endocardium minimally contributes to coronary endothelium in the embryonic ventricular free walls. CONCLUSIONS Our study suggested that SV, but not endocardium, is the major origin for coronary endothelium in the embryonic ventricular free walls. This work thus resolves the recent controversy over the developmental origin of coronary endothelium, providing the basis for studying coronary vessel formation and regeneration after injury.
Collapse
Affiliation(s)
- Hui Zhang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Wenjuan Pu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Guang Li
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Xiuzhen Huang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Lingjuan He
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Xueying Tian
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Qiaozhen Liu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Libo Zhang
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Sean M Wu
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Henry M Sucov
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.)
| | - Bin Zhou
- From the Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences (H.Z., W.P., X.H., L.H., X.T., Q.L., L.Z., B.Z.) and Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences (B.Z.), Chinese Academy of Sciences, Shanghai, China; Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (G.L., S.M.W.); Broad CIRM Center and Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles (H.M.S.); and School of Life Science and Technology, ShanghaiTech University, Shanghai, China (B.Z.).
| |
Collapse
|
45
|
Genetic lineage tracing identifies endocardial origin of liver vasculature. Nat Genet 2016; 48:537-43. [PMID: 27019112 DOI: 10.1038/ng.3536] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/04/2016] [Indexed: 02/08/2023]
Abstract
The hepatic vasculature is essential for liver development, homeostasis and regeneration, yet the developmental program of hepatic vessel formation and the embryonic origin of the liver vasculature remain unknown. Here we show in mouse that endocardial cells form a primitive vascular plexus surrounding the liver bud and subsequently contribute to a substantial portion of the liver vasculature. Using intersectional genetics, we demonstrate that the endocardium of the sinus venosus is a source for the hepatic plexus. Inhibition of endocardial angiogenesis results in reduced endocardial contribution to the liver vasculature and defects in liver organogenesis. We conclude that a substantial portion of liver vessels derives from the endocardium and shares a common developmental origin with coronary arteries.
Collapse
|