1
|
Xu S, Zhang H, Tian Y. Pericytes in hematogenous metastasis: mechanistic insights and therapeutic approaches. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01073-6. [PMID: 40392500 DOI: 10.1007/s13402-025-01073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Metastasis, the leading cause of cancer-related deaths, underscores the critical need to understand its regulatory mechanisms to improve prevention and treatment strategies for late-stage tumors. Hematogenous dissemination is a key route of metastasis. However, as the gatekeeper of vessels, the role of pericytes in hematogenous metastasis remains largely unknown. In this review, we comprehensively explore the contributions of pericytes throughout the metastatic cascade, particularly their functions that extend beyond influencing tumor angiogenesis. Pericytes should not be perceived as passive bystanders, but rather as active participants in various stages of the metastatic cascade. Pericytes-targeted therapy may provide novel insights for preventing and treating advanced-stage tumor.
Collapse
Affiliation(s)
- Shuo Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Hong Zhang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| |
Collapse
|
2
|
Wakisaka Y, Inai K, Harada G, Asagai S, Shimada E. Coronary anomalies in single ventricles: Insights from selective angiographic assessment. J Cardiol 2025:S0914-5087(25)00057-7. [PMID: 39965728 DOI: 10.1016/j.jjcc.2025.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 12/07/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Adults with congenital heart disease have a higher risk of coronary artery disease compared to the general population. However, there is limited information on coronary artery distribution in patients with a single ventricle (SV), which is important in understanding potential cardiovascular events. This study aimed to evaluate coronary artery morphology and anomalies in patients with SV based on selective coronary angiography (CAG). METHODS We performed a retrospective single-center study including 80 patients with SV [median age, 29 years (range 13-50); 54 % males] who underwent selective CAG at our institution between 2019 and 2023. Patients were classified into either single right or left ventricular (SRV and SLV) morphologies and categorized into D-, L-, and X-loops based on the rules of cardiovascular looping. Coronary artery morphology, dominance, and the abnormality of origin were evaluated. RESULTS Of the 80 SV patients, 56 had SRV and 24 had SLV. Patients with coronary artery looping surrounding the rudimentary chamber were more frequent in cases with D-loops. In patients with SV, right coronary artery dominance was observed in 70 %, and anomalous origins of coronary artery origins were found in 36.3 % of cases, with a higher frequency of L-loops. Overall, cardiovascular events were observed in patients with SV as follows: 46 % of patients had arrhythmias, 13 % had heart failure, and 8 % had thrombosis; there were no cardiovascular deaths. A single coronary artery was identified in 9 cases (11 %) of SV patients, exclusively in SRV, of which 2 cases (22 %) had arrhythmias and 1 case (11 %) had thrombus. CONCLUSION Selective CAG revealed a high prevalence of coronary artery anomalies in patients with SV, emphasizing the importance of comprehensive assessment and long-term follow-up to manage cardiovascular risk in this patient population.
Collapse
Affiliation(s)
- Yuko Wakisaka
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Heart Institute, Tokyo Women's Medical University, Tokyo, Japan
| | - Kei Inai
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Heart Institute, Tokyo Women's Medical University, Tokyo, Japan.
| | - Gen Harada
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Heart Institute, Tokyo Women's Medical University, Tokyo, Japan
| | - Seiji Asagai
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Heart Institute, Tokyo Women's Medical University, Tokyo, Japan
| | - Eriko Shimada
- Department of Pediatric Cardiology and Adult Congenital Cardiology, Heart Institute, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
3
|
Deng X, Liu H, Chen H, Yang Z, Wu Y, He L, Guo W. A gene-encoded bioprotein second harmonic generation (SHG) probe from Autographa californica nuclear polyhedrosis virus (AcMNPV) polyhedrin for live cell imaging. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2025; 54:21-32. [PMID: 39812816 DOI: 10.1007/s00249-024-01728-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 10/12/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025]
Abstract
Compared to fluorescence, second harmonic generation (SHG) has recently emerged as an excellent signal for imaging probes due to its unmatched advantages in terms of no photobleaching, no phototoxicity, no signal saturation, as well as the superior imaging accuracy with excellent avoidance of background noise. Existing SHG probes are constructed from heavy metals and are cellular exogenous, presenting with high cytotoxicity, difficult cellular uptake, and the limitation of non-heritability. We, therefore, initially propose an innovative gene-encoded bioprotein SHG probe derived from Autographa californica nuclear polyhedrosis virus (AcMNPV) polyhedrin. The primitive gene of AcMNPV polyhedrin was codon-optimized and mutated in its nuclear localization sequence to achieve cytoplasmic expression in mammalian cells. While providing strong SHG signals, this gene-modified AcMNPV (GM-AcMNPV) polyhedrin could be utilized as an SHG probe for cell imaging. Our experimental results demonstrated successful expression of GM-AcMNPV polyhedrin in the cytoplasm of HEK293T cells and bone mesenchymal stem cells (BMSCs), and verified its characteristic features as an SHG probe. Such SHG probes exhibit high biocompatibility and showed no hindering of central physiological activities such as the differentiation of stem cells. Most importantly, our SHG probes may be successfully used for imaging in living cells. This work will inspire the development of gene encoding-derived bioprotein SHG probes, for long-term tracing of cells/stem cells along with their division, to understand stem cell cycles, reveal stem cell-based therapy mechanisms in regenerative medicine, and unravel cell lineage origins and fates in developmental biology, among other potential applications.
Collapse
Affiliation(s)
- Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| | - Hao Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Heting Chen
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Zuojun Yang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Yuhan Wu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Li He
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
- Guangdong Provincial Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China
| | - Wenjing Guo
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| |
Collapse
|
4
|
Lv Z, Liu Z, Liu K, Lin X, Pu W, Li Y, Zhao H, Xi Y, Sui P, Vaughan AE, Gillich A, Zhou B. Alveolar regeneration by airway secretory-cell-derived p63 + progenitors. Cell Stem Cell 2024; 31:1685-1700.e6. [PMID: 39232560 DOI: 10.1016/j.stem.2024.08.005] [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: 05/07/2023] [Revised: 03/12/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
Lung injury activates epithelial stem or progenitor cells for alveolar repair and regeneration. Unraveling the origin and fate of injury-induced progenitors is crucial for elucidating lung repair mechanisms. Here, we report that p63-expressing progenitors emerge upon bleomycin-induced mouse lung injury. Single-cell RNA sequencing and clonal analysis reveal that these p63+ progenitors proliferate rapidly and differentiate into alveolar type 1 and type 2 cells through different trajectories. Dual recombinase-mediated sequential genetic-lineage tracing demonstrates that p63+ progenitors originate from airway secretory cells and subsequently generate alveolar cells. Functionally, p63 activation is essential for efficient alveolar regeneration from secretory cells post injury. Our study identifies secretory-cell-derived p63+ progenitors as contributors to alveolar repair, suggesting a potential therapeutic avenue for lung regeneration following injury.
Collapse
Affiliation(s)
- Zan Lv
- 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 200031, China
| | - Zixin Liu
- 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 200031, China
| | - Kuo Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Xiuyu Lin
- 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 200031, China
| | - Wenjuan Pu
- 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 200031, China
| | - Yan Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Huan Zhao
- 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 200031, China
| | - Ying Xi
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Pengfei Sui
- 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 200031, China
| | - Andrew E Vaughan
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Astrid Gillich
- Calico Life Sciences LLC, South San Francisco, CA 94080, USA
| | - Bin Zhou
- 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 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 310024, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
5
|
Sedmera D, Olejnickova V, Sankova B, Kolesova H, Bartos M, Kvasilova A, Phillips LC, Bamforth SD, Phillips HM. Morphological, electrophysiological, and molecular alterations in foetal noncompacted cardiomyopathy induced by disruption of ROCK signalling. Front Cell Dev Biol 2024; 12:1471751. [PMID: 39435333 PMCID: PMC11491540 DOI: 10.3389/fcell.2024.1471751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Left ventricular noncompaction cardiomyopathy is associated with heart failure, arrhythmia, and sudden cardiac death. The developmental mechanism underpinning noncompaction in the adult heart is still not fully understood, with lack of trabeculae compaction, hypertrabeculation, and loss of proliferation cited as possible causes. To study this, we utilised a mouse model of aberrant Rho kinase (ROCK) signalling in cardiomyocytes, which led to a noncompaction phenotype during embryogenesis, and monitored how this progressed after birth and into adulthood. The cause of the early noncompaction at E15.5 was attributed to a decrease in proliferation in the developing ventricular wall. By E18.5, the phenotype became patchy, with regions of noncompaction interspersed with thick compacted areas of ventricular wall. To study how this altered myoarchitecture of the heart influenced impulse propagation in the developing and adult heart, we used histology with immunohistochemistry for gap junction protein expression, optical mapping, and electrocardiography. At the prenatal stages, a clear reduction in left ventricular wall thickness, accompanied by abnormal conduction of the ectopically paced beat in that area, was observed in mutant hearts. This correlated with increased expression of connexin-40 and connexin-43 in noncompacted trabeculae. In postnatal stages, left ventricular noncompaction was resolved, but the right ventricular wall remained structurally abnormal through to adulthood with cardiomyocyte hypertrophy and retention of myocardial crypts. Thus, this is a novel model of self-correcting embryonic hypertrabeculation cardiomyopathy, but it highlights that remodelling potential differs between the left and right ventricles. We conclude that disruption of ROCK signalling induces both morphological and electrophysiological changes that evolve over time, highlighting the link between myocyte proliferation and noncompaction phenotypes and electrophysiological differentiation.
Collapse
Affiliation(s)
- David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Veronika Olejnickova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Barbora Sankova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Hana Kolesova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Martin Bartos
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Institute of Dental Medicine, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Alena Kvasilova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Lauren C. Phillips
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon D. Bamforth
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen M. Phillips
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
6
|
Hough RF, Alvira CM, Bastarache JA, Erzurum SC, Kuebler WM, Schmidt EP, Shimoda LA, Abman SH, Alvarez DF, Belvitch P, Bhattacharya J, Birukov KG, Chan SY, Cornfield DN, Dudek SM, Garcia JGN, Harrington EO, Hsia CCW, Islam MN, Jonigk DD, Kalinichenko VV, Kolb TM, Lee JY, Mammoto A, Mehta D, Rounds S, Schupp JC, Shaver CM, Suresh K, Tambe DT, Ventetuolo CE, Yoder MC, Stevens T, Damarla M. Studying the Pulmonary Endothelium in Health and Disease: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 71:388-406. [PMID: 39189891 PMCID: PMC11450313 DOI: 10.1165/rcmb.2024-0330st] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Lung endothelium resides at the interface between the circulation and the underlying tissue, where it senses biochemical and mechanical properties of both the blood as it flows through the vascular circuit and the vessel wall. The endothelium performs the bidirectional signaling between the blood and tissue compartments that is necessary to maintain homeostasis while physically separating both, facilitating a tightly regulated exchange of water, solutes, cells, and signals. Disruption in endothelial function contributes to vascular disease, which can manifest in discrete vascular locations along the artery-to-capillary-to-vein axis. Although our understanding of mechanisms that contribute to endothelial cell injury and repair in acute and chronic vascular disease have advanced, pathophysiological mechanisms that underlie site-specific vascular disease remain incompletely understood. In an effort to improve the translatability of mechanistic studies of the endothelium, the American Thoracic Society convened a workshop to optimize rigor, reproducibility, and translation of discovery to advance our understanding of endothelial cell function in health and disease.
Collapse
|
7
|
Jeong JY, Bafor AE, Freeman BH, Chen PR, Park ES, Kim E. Pathophysiology in Brain Arteriovenous Malformations: Focus on Endothelial Dysfunctions and Endothelial-to-Mesenchymal Transition. Biomedicines 2024; 12:1795. [PMID: 39200259 PMCID: PMC11351371 DOI: 10.3390/biomedicines12081795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
Brain arteriovenous malformations (bAVMs) substantially increase the risk for intracerebral hemorrhage (ICH), which is associated with significant morbidity and mortality. However, the treatment options for bAVMs are severely limited, primarily relying on invasive methods that carry their own risks for intraoperative hemorrhage or even death. Currently, there are no pharmaceutical agents shown to treat this condition, primarily due to a poor understanding of bAVM pathophysiology. For the last decade, bAVM research has made significant advances, including the identification of novel genetic mutations and relevant signaling in bAVM development. However, bAVM pathophysiology is still largely unclear. Further investigation is required to understand the detailed cellular and molecular mechanisms involved, which will enable the development of safer and more effective treatment options. Endothelial cells (ECs), the cells that line the vascular lumen, are integral to the pathogenesis of bAVMs. Understanding the fundamental role of ECs in pathological conditions is crucial to unraveling bAVM pathophysiology. This review focuses on the current knowledge of bAVM-relevant signaling pathways and dysfunctions in ECs, particularly the endothelial-to-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
| | | | | | | | | | - Eunhee Kim
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.Y.J.); (A.E.B.); (B.H.F.); (P.R.C.); (E.S.P.)
| |
Collapse
|
8
|
Foglio E, D'Avorio E, Nieri R, Russo MA, Limana F. Epicardial EMT and cardiac repair: an update. Stem Cell Res Ther 2024; 15:219. [PMID: 39026298 PMCID: PMC11264588 DOI: 10.1186/s13287-024-03823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/30/2024] [Indexed: 07/20/2024] Open
Abstract
Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, Latina, Italy
| | - Erica D'Avorio
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy
| | - Riccardo Nieri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Federica Limana
- Dipartimento di Promozione delle Scienze Umane e della Qualità della Vita, San Raffaele University of Rome, Rome, Italy.
- Laboratorio di Patologia Cellulare e Molecolare, IRCCS San Raffaele Roma, Rome, Italy.
| |
Collapse
|
9
|
Sultan I, Ramste M, Peletier P, Hemanthakumar KA, Ramanujam D, Tirronen A, von Wright Y, Antila S, Saharinen P, Eklund L, Mervaala E, Ylä-Herttuala S, Engelhardt S, Kivelä R, Alitalo K. Contribution of VEGF-B-Induced Endocardial Endothelial Cell Lineage in Physiological Versus Pathological Cardiac Hypertrophy. Circ Res 2024; 134:1465-1482. [PMID: 38655691 PMCID: PMC11542978 DOI: 10.1161/circresaha.123.324136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/19/2024] [Accepted: 04/08/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Preclinical studies have shown the therapeutic potential of VEGF-B (vascular endothelial growth factor B) in revascularization of the ischemic myocardium, but the associated cardiac hypertrophy and adverse side effects remain a concern. To understand the importance of endothelial proliferation and migration for the beneficial versus adverse effects of VEGF-B in the heart, we explored the cardiac effects of autocrine versus paracrine VEGF-B expression in transgenic and gene-transduced mice. METHODS We used single-cell RNA sequencing to compare cardiac endothelial gene expression in VEGF-B transgenic mouse models. Lineage tracing was used to identify the origin of a VEGF-B-induced novel endothelial cell population and adeno-associated virus-mediated gene delivery to compare the effects of VEGF-B isoforms. Cardiac function was investigated using echocardiography, magnetic resonance imaging, and micro-computed tomography. RESULTS Unlike in physiological cardiac hypertrophy driven by a cardiomyocyte-specific VEGF-B transgene (myosin heavy chain alpha-VEGF-B), autocrine VEGF-B expression in cardiac endothelium (aP2 [adipocyte protein 2]-VEGF-B) was associated with septal defects and failure to increase perfused subendocardial capillaries postnatally. Paracrine VEGF-B led to robust proliferation and myocardial migration of a novel cardiac endothelial cell lineage (VEGF-B-induced endothelial cells) of endocardial origin, whereas autocrine VEGF-B increased proliferation of VEGF-B-induced endothelial cells but failed to promote their migration and efficient contribution to myocardial capillaries. The surviving aP2-VEGF-B offspring showed an altered ratio of secreted VEGF-B isoforms and developed massive pathological cardiac hypertrophy with a distinct cardiac vessel pattern. In the normal heart, we found a small VEGF-B-induced endothelial cell population that was only minimally expanded during myocardial infarction but not during physiological cardiac hypertrophy associated with mouse pregnancy. CONCLUSIONS Paracrine and autocrine secretions of VEGF-B induce expansion of a specific endocardium-derived endothelial cell population with distinct angiogenic markers. However, autocrine VEGF-B signaling fails to promote VEGF-B-induced endothelial cell migration and contribution to myocardial capillaries, predisposing to septal defects and inducing a mismatch between angiogenesis and myocardial growth, which results in pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ibrahim Sultan
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Markus Ramste
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Pim Peletier
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Karthik Amudhala Hemanthakumar
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Deepak Ramanujam
- Institute of Pharmacology and Toxicology, Technical University of Munich, DZHK partner site Munich Heart Alliance, Germany (D.R., S.E.)
- RNATICS GmbH, Planegg, Germany (D.R.)
| | - Annakaisa Tirronen
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (A.T., S.Y.-H.)
| | - Ylva von Wright
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Salli Antila
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Pipsa Saharinen
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Finland (L.E.)
| | - Eero Mervaala
- Department of Pharmacology (E.M.), Faculty of Medicine, University of Helsinki, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland, Kuopio, Finland (A.T., S.Y.-H.)
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich, DZHK partner site Munich Heart Alliance, Germany (D.R., S.E.)
| | - Riikka Kivelä
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Stem Cells and Metabolism Research Program (R.K.), Faculty of Medicine, University of Helsinki, Finland
- Faculty of Sport and Health Sciences, University of Jyväskylä, Finland (R.K.)
| | - Kari Alitalo
- Wihuri Research Institute (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., R.K., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
- Translational Cancer Medicine Program (I.S., M.R., P.P., K.A.H., Y.v.W., S.A., P.S., K.A.), Faculty of Medicine, Biomedicum Helsinki, University of Helsinki, Finland
| |
Collapse
|
10
|
Sullivan JM, Bagnell AM, Alevy J, Avila EM, Mihaljević L, Saavedra-Rivera PC, Kong L, Huh JS, McCray BA, Aisenberg WH, Zuberi AR, Bogdanik L, Lutz CM, Qiu Z, Quinlan KA, Searson PC, Sumner CJ. Gain-of-function mutations of TRPV4 acting in endothelial cells drive blood-CNS barrier breakdown and motor neuron degeneration in mice. Sci Transl Med 2024; 16:eadk1358. [PMID: 38776392 PMCID: PMC11316273 DOI: 10.1126/scitranslmed.adk1358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Blood-CNS barrier disruption is a hallmark of numerous neurological disorders, yet whether barrier breakdown is sufficient to trigger neurodegenerative disease remains unresolved. Therapeutic strategies to mitigate barrier hyperpermeability are also limited. Dominant missense mutations of the cation channel transient receptor potential vanilloid 4 (TRPV4) cause forms of hereditary motor neuron disease. To gain insights into the cellular basis of these disorders, we generated knock-in mouse models of TRPV4 channelopathy by introducing two disease-causing mutations (R269C and R232C) into the endogenous mouse Trpv4 gene. TRPV4 mutant mice exhibited weakness, early lethality, and regional motor neuron loss. Genetic deletion of the mutant Trpv4 allele from endothelial cells (but not neurons, glia, or muscle) rescued these phenotypes. Symptomatic mutant mice exhibited focal disruptions of blood-spinal cord barrier (BSCB) integrity, associated with a gain of function of mutant TRPV4 channel activity in neural vascular endothelial cells (NVECs) and alterations of NVEC tight junction structure. Systemic administration of a TRPV4-specific antagonist abrogated channel-mediated BSCB impairments and provided a marked phenotypic rescue of symptomatic mutant mice. Together, our findings show that mutant TRPV4 channels can drive motor neuron degeneration in a non-cell autonomous manner by precipitating focal breakdown of the BSCB. Further, these data highlight the reversibility of TRPV4-mediated BSCB impairments and identify a potential therapeutic strategy for patients with TRPV4 mutations.
Collapse
Affiliation(s)
- Jeremy M. Sullivan
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Anna M. Bagnell
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Jonathan Alevy
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Elvia Mena Avila
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island; Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island; Kingston, RI 02881, USA
| | - Ljubica Mihaljević
- Department of Physiology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | | | - Lingling Kong
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Jennifer S. Huh
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Brett A. McCray
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - William H. Aisenberg
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | | | | | | | - Zhaozhu Qiu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
- Department of Physiology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| | - Katharina A. Quinlan
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island; Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island; Kingston, RI 02881, USA
| | - Peter C. Searson
- Institute for Nanobiotechnology, Johns Hopkins University; Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University; Baltimore, MD 21218, USA
| | - Charlotte J. Sumner
- Department of Neurology, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine; Baltimore, MD 21205, USA
| |
Collapse
|
11
|
McCracken IR, Smart N. Control of coronary vascular cell fate in development and regeneration. Semin Cell Dev Biol 2024; 155:50-61. [PMID: 37714806 DOI: 10.1016/j.semcdb.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/17/2023]
Abstract
The coronary vasculature consists of a complex hierarchal network of arteries, veins, and capillaries which collectively function to perfuse the myocardium. However, the pathways controlling the temporally and spatially restricted mechanisms underlying the formation of this vascular network remain poorly understood. In recent years, the increasing use and refinement of transgenic mouse models has played an instrumental role in offering new insights into the cellular origins of the coronary vasculature, as well as identifying a continuum of transitioning cell states preceding the full maturation of the coronary vasculature. Coupled with the emergence of single cell RNA sequencing platforms, these technologies have begun to uncover the key regulatory factors mediating the convergence of distinct cellular origins to ensure the formation of a collectively functional, yet phenotypically diverse, vascular network. Furthermore, improved understanding of the key regulatory factors governing coronary vessel formation in the embryo may provide crucial clues into future therapeutic strategies to reactivate these developmentally functional mechanisms to drive the revascularisation of the ischaemic adult heart.
Collapse
Affiliation(s)
- Ian R McCracken
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, United Kingdom
| | - Nicola Smart
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX3 7TY, United Kingdom.
| |
Collapse
|
12
|
Zhang M, Lui KO, Zhou B. Application of New Lineage Tracing Techniques in Cardiovascular Development and Physiology. Circ Res 2024; 134:445-458. [PMID: 38359092 DOI: 10.1161/circresaha.123.323179] [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] [Indexed: 02/17/2024]
Abstract
Cardiovascular disease has been the leading cause of mortality and morbidity worldwide in the past 3 decades. Multiple cell lineages undergo dynamic alternations in gene expression, cell state determination, and cell fate conversion to contribute, adapt, and even modulate the pathophysiological processes during disease progression. There is an urgent need to understand the intricate cellular and molecular underpinnings of cardiovascular cell development in homeostasis and pathogenesis. Recent strides in lineage tracing methodologies have revolutionized our understanding of cardiovascular biology with the identification of new cellular origins, fates, plasticity, and heterogeneity within the cardiomyocyte, endothelial, and mesenchymal cell populations. In this review, we introduce the new technologies for lineage tracing of cardiovascular cells and summarize their applications in studying cardiovascular development, diseases, repair, and regeneration.
Collapse
Affiliation(s)
- MingJun Zhang
- New Cornerstone Investigator Institute, 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, China (M.J., B.Z.)
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, China (K.O.L.)
| | - Bin Zhou
- New Cornerstone Investigator Institute, 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, China (M.J., B.Z.)
- School of Life Science and Technology, ShanghaiTech University, China (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, China (B.Z.)
| |
Collapse
|
13
|
Abstract
Permanent fibrosis and chronic deterioration of heart function in patients after myocardial infarction present a major health-care burden worldwide. In contrast to the restricted potential for cellular and functional regeneration of the adult mammalian heart, a robust capacity for cardiac regeneration is seen during the neonatal period in mammals as well as in the adults of many fish and amphibian species. However, we lack a complete understanding as to why cardiac regeneration takes place more efficiently in some species than in others. The capacity of the heart to regenerate after injury is controlled by a complex network of cellular and molecular mechanisms that form a regulatory landscape, either permitting or restricting regeneration. In this Review, we provide an overview of the diverse array of vertebrates that have been studied for their cardiac regenerative potential and discuss differential heart regeneration outcomes in closely related species. Additionally, we summarize current knowledge about the core mechanisms that regulate cardiac regeneration across vertebrate species.
Collapse
Affiliation(s)
- Michael Weinberger
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK
- MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Paul R Riley
- Institute of Developmental & Regenerative Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
14
|
Nguyen TM, Geng X, Wei Y, Ye L, Garry DJ, Zhang J. Single-cell RNA sequencing analysis identifies one subpopulation of endothelial cells that proliferates and another that undergoes the endothelial-mesenchymal transition in regenerating pig hearts. Front Bioeng Biotechnol 2024; 11:1257669. [PMID: 38288246 PMCID: PMC10823534 DOI: 10.3389/fbioe.2023.1257669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 12/04/2023] [Indexed: 01/31/2024] Open
Abstract
Background: In our previous work, we demonstrated that when newborn pigs undergo apical resection (AR) on postnatal day 1 (P1), the animals' hearts were completely recover from a myocardial infarction (MI) that occurs on postnatal day 28 (P28); single-nucleus RNA sequencing (snRNAseq) data suggested that this recovery was achieved by regeneration of pig cardiomyocyte subpopulations in response to MI. However, coronary vasculature also has a key role in promoting cardiac repair. Method: Thus, in this report, we used autoencoder algorithms to analyze snRNAseq data from endothelial cells (ECs) in the hearts of the same animals. Main results: Our results identified five EC clusters, three composed of vascular ECs (VEC1-3) and two containing lymphatic ECs (LEC1-2). Cells from VEC1 expressed elevated levels of each of five cell-cyclespecific markers (Aurora Kinase B [AURKB], Marker of Proliferation Ki-67 [MKI67], Inner Centromere Protein [INCENP], Survivin [BIRC5], and Borealin [CDCA8]), as well as a number of transcription factors that promote EC proliferation, while (VEC3 was enriched for genes that regulate intercellular junctions, participate in transforming growth factor β (TGFβ), bone morphogenic protein (BMP) signaling, and promote the endothelial mesenchymal transition (EndMT). The remaining VEC2 did not appear to participate directly in the angiogenic response to MI, but trajectory analyses indicated that it may serve as a reservoir for the generation of VEC1 and VEC3 ECs in response to MI. Notably, only the VEC3 cluster was more populous in regenerating (i.e., ARP1MIP28) than non-regenerating (i.e., MIP28) hearts during the 1-week period after MI induction, which suggests that further investigation of the VEC3 cluster could identify new targets for improving myocardial recovery after MI. Histological analysis of KI67 and EndMT marker PDGFRA demonstrated that while the expression of proliferation of endothelial cells was not significantly different, expression of EndMT markers was significantly higher among endothelial cells of ARP1MIP28 hearts compared to MIP28 hearts, which were consistent with snRNAseq analysis of clusters VEC1 and VEC3. Furthermore, upregulated secrete genes by VEC3 may promote cardiomyocyte proliferation via the Pi3k-Akt and ERBB signaling pathways, which directly contribute to cardiac muscle regeneration. Conclusion: In regenerative heart, endothelial cells may express EndMT markers, and this process could contribute to regeneration via a endothelial-cardiomyocyte crosstalk that supports cardiomyocyte proliferation.
Collapse
Affiliation(s)
- Thanh Minh Nguyen
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Xiaoxiao Geng
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yuhua Wei
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Daniel J. Garry
- Department of Medicine, School of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, Cardiovascular Diseases, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
15
|
Ruiz-Villalba A, Guadix JA, Pérez-Pomares JM. Epicardium and Coronary Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:155-166. [PMID: 38884710 DOI: 10.1007/978-3-031-44087-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Congenital anomalies and acquired diseases of the coronary blood vessels are of great clinical relevance. The early diagnosis of these conditions remains, however, challenging. In order to improve our knowledge of these ailments, progress has to be achieved in the research of the molecular and cellular mechanisms that control development of the coronary vascular bed. The aim of this chapter is to provide a succint account of the key elements of coronary blood vessel development, especially in the context of the role played by the epicardium and epicardial cellular derivatives. We will discuss the importance of the epicardium in coronary blood vessel morphogenesis, from the contribution of the epicardially derived mesenchyme to these blood vessels to its role as an instructive signaling center, attempting to relate these concepts to the origin of coronary disease.
Collapse
Affiliation(s)
- Adrián Ruiz-Villalba
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Campanillas (Málaga), Spain
| | - Juan Antonio Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Campanillas (Málaga), Spain
| | - José M Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain.
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Campanillas (Málaga), Spain.
| |
Collapse
|
16
|
Abstract
Cardiac fibrosis, which is the buildup of proteins in the connective tissues of the heart, can lead to end-stage extracellular matrix (ECM) remodeling and ultimately heart failure. Cardiac remodeling involves changes in gene expression in cardiac cells and ECM, which significantly leads to the morbidity and mortality in heart failure. However, despite extensive research, the elusive intricacies underlying cardiac fibrosis remain unidentified. Periostin, an extracellular matrix (ECM) protein of the fasciclin superfamily, acts as a scaffold for building complex architectures in the ECM, which improves intermolecular interactions and augments the mechanical properties of connective tissues. Recent research has shown that periostin not only contributes to normal ECM homeostasis in a healthy heart but also serves as a potent inducible regulator of cellular reorganization in cardiac fibrosis. Here, we reviewed the constitutive domain of periostin and its interaction with other ECM proteins. We have also discussed the critical pathophysiological functions of periostin in cardiac remodeling mechanisms, including two distinct yet potentially intertwined mechanisms. Furthermore, we will focus on the intrinsic complexities within periostin research, particularly surrounding the contentious issues observed in experimental findings.
Collapse
Affiliation(s)
- Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Bailu Wang
- Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Clinical Research Center for Emergency and Critical Care Medicine of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
17
|
Guadix JA, Ruiz-Villalba A, Pérez-Pomares JM. Congenital Coronary Blood Vessel Anomalies: Animal Models and the Integration of Developmental Mechanisms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1441:817-831. [PMID: 38884751 DOI: 10.1007/978-3-031-44087-8_49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Coronary blood vessels are in charge of sustaining cardiac homeostasis. It is thus logical that coronary congenital anomalies (CCA) directly or indirectly associate with multiple cardiac conditions, including sudden death. The coronary vascular system is a sophisticated, highly patterned anatomical entity, and therefore a wide range of congenital malformations of the coronary vasculature have been described. Despite the clinical interest of CCA, very few attempts have been made to relate specific embryonic developmental mechanisms to the congenital anomalies of these blood vessels. This is so because developmental data on the morphogenesis of the coronary vascular system derive from complex studies carried out in animals (mostly transgenic mice), and are not often accessible to the clinician, who, in turn, possesses essential information on the significance of CCA. During the last decade, advances in our understanding of normal embryonic development of coronary blood vessels have provided insight into the cellular and molecular mechanisms underlying coronary arteries anomalies. These findings are the base for our attempt to offer plausible embryological explanations to a variety of CCA as based on the analysis of multiple animal models for the study of cardiac embryogenesis, and present them in an organized manner, offering to the reader developmental mechanistic explanations for the pathogenesis of these anomalies.
Collapse
Affiliation(s)
- Juan Antonio Guadix
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
| | - Adrián Ruiz-Villalba
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain
| | - José M Pérez-Pomares
- Department of Animal Biology, Faculty of Sciences, University of Málaga, Málaga, Spain.
- Instituto de Biomedicina de Málaga (IBIMA)-Plataforma BIONAND, Málaga, Spain.
| |
Collapse
|
18
|
Sui Y, Kou S, Ge K, Cao J, Liu C, Zhang H. Expression analysis of plvap in mouse heart development, homeostasis and injury. Gene Expr Patterns 2023; 50:119343. [PMID: 37774966 DOI: 10.1016/j.gep.2023.119343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 09/12/2023] [Accepted: 09/27/2023] [Indexed: 10/01/2023]
Abstract
Plasmalemma vesicle associated protein (PLVAP) is commonly considered to be specifically expressed in endothelial cells in which it localized to diaphragms of caveolae, fenestrae, and transendothelial channels. PLVAP is reported to be an important regulator of heart development and a novel target to promote cardiac repair in the ischemic heart. However, the dynamics of plvap expression in heart development, homeostasis and pathology have not been comprehensively described. In this study, we analyzed the temporal and spatial expression of plvap in mouse heart under different conditions. We found that, during embryonic and neonatal stages, PLVAP was detected in endocardial endothelial cells, epicardial mesothelial cells, and a small amount of coronary vascular endothelial cells. In adult heart, PLVAP was also identified in endocardial cells and a few coronary vascular endothelial cells. However, epicardial expression of PLVAP was lost during postnatal heart development and cannot be detected in mouse heart by immunostaining since 3-week-old. We also analyzed the expression of plvap in a model of cardiac hypertrophy and failure induced by transverse aortic constriction surgery, and identified expression of PLVAP in endocardial cells and coronary vascular endothelial cells in the injured heart. This study provides new evidence to better understand the role of plvap in mouse heart development and injury.
Collapse
Affiliation(s)
- Yu Sui
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shan Kou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kaixin Ge
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinjun Cao
- Department of Pediatric Critical Care Medicine, Xinhua Hospital, Affiliated to the Medical School of Shanghai Jiao Tong University, Shanghai, China.
| | - Chen Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Hui Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
19
|
Zhan R, Meng X, Tian D, Xu J, Cui H, Yang J, Xu Y, Shi M, Xue J, Yu W, Hu G, Li K, Ge X, Zhang Q, Zhao M, Du J, Guo X, Xu W, Gao Y, Yao C, Chen F, Chen Y, Shan W, Zhu Y, Ji L, Pan B, Yu Y, Li W, Zhao X, He Q, Liu X, Huang Y, Liao S, Zhou B, Chui D, Chen YE, Sun Z, Dong E, Wang Y, Zheng L. NAD + rescues aging-induced blood-brain barrier damage via the CX43-PARP1 axis. Neuron 2023; 111:3634-3649.e7. [PMID: 37683629 DOI: 10.1016/j.neuron.2023.08.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 04/17/2023] [Accepted: 08/09/2023] [Indexed: 09/10/2023]
Abstract
Blood-brain barrier (BBB) function deteriorates during aging, contributing to cognitive impairment and neurodegeneration. It is unclear what drives BBB leakage in aging and how it can be prevented. Using single-nucleus transcriptomics, we identified decreased connexin 43 (CX43) expression in cadherin-5+ (Cdh5+) cerebral vascular cells in naturally aging mice and confirmed it in human brain samples. Global or Cdh5+ cell-specific CX43 deletion in mice exacerbated BBB dysfunction during aging. The CX43-dependent effect was not due to its canonical gap junction function but was associated with reduced NAD+ levels and mitochondrial dysfunction through NAD+-dependent sirtuin 3 (SIRT3). CX43 interacts with and negatively regulates poly(ADP-ribose) polymerase 1 (PARP1). Pharmacologic inhibition of PARP1 by olaparib or nicotinamide mononucleotide (NMN) supplementation rescued NAD+ levels and alleviated aging-associated BBB leakage. These findings establish the endothelial CX43-PARP1-NAD+ pathway's role in vascular aging and identify a potential therapeutic strategy to combat aging-associated BBB leakage with neuroprotective implications.
Collapse
Affiliation(s)
- Rui Zhan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Xia Meng
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Dongping Tian
- Department of Pathology, Medical College, Shantou University, Shantou, China
| | - Jie Xu
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Hongtu Cui
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Jialei Yang
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Yangkai Xu
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Mingming Shi
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Jing Xue
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Weiwei Yu
- Peking University Shenzhen Hospital, Beijing, China
| | - Gaofei Hu
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Ke Li
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Xiaoxiao Ge
- Beijing Institute Brain Disorders, Capital Medical University, Beijing, China
| | - Qi Zhang
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Jianyong Du
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Xin Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Wenli Xu
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Yang Gao
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| | - Changyu Yao
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Fan Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yue Chen
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Wenxin Shan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Yujie Zhu
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Bing Pan
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Yan Yu
- Chinese Institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, China
| | - Wenguang Li
- Institute of Genetics and Development Biology, Chinese Academy of Sciences, Beijing, China
| | - Xuyang Zhao
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China
| | - Qihua He
- Center of Medical and Health Analysis, Peking University, Beijing, China
| | - Xiaohui Liu
- National Protein Science Technology Center, Tsinghua University, Beijing, China
| | - Yue Huang
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China
| | - Shengyou Liao
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University, Shenzhen People's Hospital, Shenzhen, 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, Chinese Academy of Sciences, Shanghai, China
| | - Dehua Chui
- Beijing Key Laboratory of Magnetic Resonance Imaging Devices and Technology and Department of Neurology, Peking University Third Hospital, Beijing, China
| | - Y Eugene Chen
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, USA
| | - Zheng Sun
- Department of Medicine and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Erdan Dong
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing, China.
| | - Yongjun Wang
- Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, Peking University, Beijing 100191, China; Beijing Tiantan Hospital, China National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, Beijing, China; The Institute of Systems Biomedicine, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China.
| |
Collapse
|
20
|
Liu C, Yang F, Su X, Zhang Z, Xing Y. ScRNA-seq and spatial transcriptomics: exploring the occurrence and treatment of coronary-related diseases starting from development. Front Cardiovasc Med 2023; 10:1064949. [PMID: 37416923 PMCID: PMC10319627 DOI: 10.3389/fcvm.2023.1064949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 05/22/2023] [Indexed: 07/08/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is a new technology that can be used to explore molecular changes in complex cell clusters at the single-cell level. Single-cell spatial transcriptomic technology complements the cell-space location information lost during single-cell sequencing. Coronary artery disease is an important cardiovascular disease with high mortality rates. Many studies have explored the physiological development and pathological changes in coronary arteries from the perspective of single cells using single-cell spatial transcriptomic technology. This article reviews the molecular mechanisms underlying coronary artery development and diseases as revealed by scRNA-seq combined with spatial transcriptomic technology. Based on these mechanisms, we discuss the possible new treatments for coronary diseases.
Collapse
|
21
|
Lu P, Wu B, Wang Y, Russell M, Liu Y, Bernard DJ, Zheng D, Zhou B. Prerequisite endocardial-mesenchymal transition for murine cardiac trabecular angiogenesis. Dev Cell 2023; 58:791-805.e4. [PMID: 37023750 PMCID: PMC10656710 DOI: 10.1016/j.devcel.2023.03.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/01/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023]
Abstract
Coronary heart disease damages the trabecular myocardium, and the regeneration of trabecular vessels may alleviate ischemic injury. However, the origins and developmental mechanisms of trabecular vessels remain unknown. Here, we show that murine ventricular endocardial cells generate trabecular vessels through an "angioEMT" mechanism. Time course fate mapping defined a specific wave of trabecular vascularization by ventricular endocardial cells. Single-cell transcriptomics and immunofluorescence identified a subpopulation of ventricular endocardial cells that underwent endocardial-mesenchymal transition (EMT) before these cells generated trabecular vessels. Ex vivo pharmacological activation and in vivo genetic inactivation experiments identified an EMT signal in ventricular endocardial cells involving SNAI2-TGFB2/TGFBR3, which was a prerequisite for later trabecular-vessel formation. Additional loss- and gain-of-function genetic studies showed that VEGFA-NOTCH1 signaling regulated post-EMT trabecular angiogenesis by ventricular endocardial cells. Our finding that trabecular vessels originate from ventricular endocardial cells through a two-step angioEMT mechanism could inform better regeneration medicine for coronary heart disease.
Collapse
Affiliation(s)
- Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China.
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Yidong Wang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Cardiovascular Research Center, School of Basic Medical Sciences, Jiaotong University, Xi'an 710061, China
| | - Megan Russell
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Departments of Pediatrics and Medicine, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA.
| |
Collapse
|
22
|
Abstract
Vascular endothelial cells form the inner layer of blood vessels where they have a key role in the development and maintenance of the functional circulatory system and provide paracrine support to surrounding non-vascular cells. Technical advances in the past 5 years in single-cell genomics and in in vivo genetic labelling have facilitated greater insights into endothelial cell development, plasticity and heterogeneity. These advances have also contributed to a new understanding of the timing of endothelial cell subtype differentiation and its relationship to the cell cycle. Identification of novel tissue-specific gene expression patterns in endothelial cells has led to the discovery of crucial signalling pathways and new interactions with other cell types that have key roles in both tissue maintenance and disease pathology. In this Review, we describe the latest findings in vascular endothelial cell development and diversity, which are often supported by large-scale, single-cell studies, and discuss the implications of these findings for vascular medicine. In addition, we highlight how techniques such as single-cell multimodal omics, which have become increasingly sophisticated over the past 2 years, are being utilized to study normal vascular physiology as well as functional perturbations in disease.
Collapse
Affiliation(s)
- Emily Trimm
- Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
Palmquist-Gomes P, Ruiz-Villalba A, Guadix JA, Romero JP, Bessiéres B, MacGrogan D, Conejo L, Ortiz A, Picazo B, Houyel L, Gómez-Cabrero D, Meilhac SM, de la Pompa JL, Pérez-Pomares JM. Origin of congenital coronary arterio-ventricular fistulae from anomalous epicardial and myocardial development. Exp Mol Med 2023; 55:228-239. [PMID: 36653444 PMCID: PMC9898521 DOI: 10.1038/s12276-022-00913-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 01/19/2023] Open
Abstract
Coronary Artery Fistulae (CAFs) are cardiac congenital anomalies consisting of an abnormal communication of a coronary artery with either a cardiac chamber or another cardiac vessel. In humans, these congenital anomalies can lead to complications such as myocardial hypertrophy, endocarditis, heart dilatation, and failure. Unfortunately, despite their clinical relevance, the aetiology of CAFs remains unknown. In this work, we have used two different species (mouse and avian embryos) to experimentally model CAFs morphogenesis. Both conditional Itga4 (alpha 4 integrin) epicardial deletion in mice and cryocauterisation of chick embryonic hearts disrupted epicardial development and ventricular wall growth, two essential events in coronary embryogenesis. Our results suggest that myocardial discontinuities in the embryonic ventricular wall promote the early contact of the endocardium with epicardial-derived coronary progenitors at the cardiac surface, leading to ventricular endocardial extrusion, precocious differentiation of coronary smooth muscle cells, and the formation of pouch-like aberrant coronary-like structures in direct connection with the ventricular lumen. The structure of these CAF-like anomalies was compared with histopathological data from a human CAF. Our results provide relevant information for the early diagnosis of these congenital anomalies and the molecular mechanisms that regulate their embryogenesis.
Collapse
Affiliation(s)
- P. Palmquist-Gomes
- grid.10215.370000 0001 2298 7828Department of Animal Biology, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain ,grid.10215.370000 0001 2298 7828IBIMA-Plataforma BIONAND (Junta de Andalucía, Universidad de Málaga), 29590 Campanillas (Málaga), Málaga, Spain ,Université de Paris, Imagine-Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, 75015 Paris, France
| | - A. Ruiz-Villalba
- grid.10215.370000 0001 2298 7828Department of Animal Biology, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain ,grid.10215.370000 0001 2298 7828IBIMA-Plataforma BIONAND (Junta de Andalucía, Universidad de Málaga), 29590 Campanillas (Málaga), Málaga, Spain
| | - J. A. Guadix
- grid.10215.370000 0001 2298 7828Department of Animal Biology, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain ,grid.10215.370000 0001 2298 7828IBIMA-Plataforma BIONAND (Junta de Andalucía, Universidad de Málaga), 29590 Campanillas (Málaga), Málaga, Spain
| | - J. P. Romero
- grid.5924.a0000000419370271Advanced Genomics Laboratory, Program of Hemato-Oncology, CIMA, University of Navarra, Pamplona, 31008 Spain
| | - B. Bessiéres
- Université de Paris, M3C-Necker Enfants malades, AP-HP, 75015 Paris, France
| | - D. MacGrogan
- grid.413448.e0000 0000 9314 1427Intercellular Signalling in Cardiovascular Development and Disease Laboratory, National Centre of Cardiovascular Research-Instituto de Salud Carlos III, 28029 Madrid, Spain ,grid.510932.cCIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - L. Conejo
- grid.411457.2Hospital Materno-Infantil de Málaga, Instituto Malagueño de Biomedicina (IBIMA), 29080 Málaga, Spain
| | - A. Ortiz
- grid.411457.2Hospital Materno-Infantil de Málaga, Instituto Malagueño de Biomedicina (IBIMA), 29080 Málaga, Spain
| | - B. Picazo
- grid.411457.2Hospital Materno-Infantil de Málaga, Instituto Malagueño de Biomedicina (IBIMA), 29080 Málaga, Spain
| | - L. Houyel
- Université de Paris, M3C-Necker Enfants malades, AP-HP, 75015 Paris, France
| | - D. Gómez-Cabrero
- Traslational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), IdiSNA, 31008 Pamplona, Spain ,grid.45672.320000 0001 1926 5090Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, 23955 Thuwal, Saudi Arabia
| | - S. M. Meilhac
- Université de Paris, Imagine-Institut Pasteur, Unit of Heart Morphogenesis, INSERM UMR1163, 75015 Paris, France
| | - J. L. de la Pompa
- grid.413448.e0000 0000 9314 1427Intercellular Signalling in Cardiovascular Development and Disease Laboratory, National Centre of Cardiovascular Research-Instituto de Salud Carlos III, 28029 Madrid, Spain ,grid.510932.cCIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - J. M. Pérez-Pomares
- grid.10215.370000 0001 2298 7828Department of Animal Biology, Faculty of Sciences, University of Málaga, 29071 Málaga, Spain ,grid.10215.370000 0001 2298 7828IBIMA-Plataforma BIONAND (Junta de Andalucía, Universidad de Málaga), 29590 Campanillas (Málaga), Málaga, Spain ,grid.510932.cCIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| |
Collapse
|
24
|
Zhang M, Pu W, Li J, Han M, Han X, Zhang Z, Lv Z, Smart N, Wang L, Zhou B. Coronary vessels contribute to de novo endocardial cells in the endocardium-depleted heart. Cell Discov 2023; 9:4. [PMID: 36627273 PMCID: PMC9832008 DOI: 10.1038/s41421-022-00486-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 10/20/2022] [Indexed: 01/12/2023] Open
Affiliation(s)
- 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
| | - 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
| | - Jie 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
| | - Maoying Han
- School of Life Science and Technology, ShanghaiTech University, 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
| | - Zhenqian 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
| | - Zan Lv
- 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
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, British Heart Foundation Centre of Regenerative Medicine, University of Oxford, Oxford, UK
| | - Lixin Wang
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Bin Zhou
- 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. .,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, China.
| |
Collapse
|
25
|
Single-cell transcriptomic analysis identifies murine heart molecular features at embryonic and neonatal stages. Nat Commun 2022; 13:7960. [PMID: 36575170 PMCID: PMC9794824 DOI: 10.1038/s41467-022-35691-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Heart development is a continuous process involving significant remodeling during embryogenesis and neonatal stages. To date, several groups have used single-cell sequencing to characterize the heart transcriptomes but failed to capture the progression of heart development at most stages. This has left gaps in understanding the contribution of each cell type across cardiac development. Here, we report the transcriptional profile of the murine heart from early embryogenesis to late neonatal stages. Through further analysis of this dataset, we identify several transcriptional features. We identify gene expression modules enriched at early embryonic and neonatal stages; multiple cell types in the left and right atriums are transcriptionally distinct at neonatal stages; many congenital heart defect-associated genes have cell type-specific expression; stage-unique ligand-receptor interactions are mostly between epicardial cells and other cell types at neonatal stages; and mutants of epicardium-expressed genes Wt1 and Tbx18 have different heart defects. Assessment of this dataset serves as an invaluable source of information for studies of heart development.
Collapse
|
26
|
Hagen MW, Louey S, Alaniz SM, Brown L, Lindner JR, Jonker SS. Coronary conductance in the normal development of sheep during the perinatal period. PHYSICS REPORTS-REVIEW SECTION OF PHYSICS LETTERS 2022; 10:e15523. [PMID: 36461657 PMCID: PMC9718948 DOI: 10.14814/phy2.15523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 12/04/2022]
Abstract
Birth is associated with substantial shifts in cardiovascular physiology. Little is known about coronary vascular adaptations during this period. We used fetal and neonatal lambs to measure coronary function at late gestation (92% of term) and shortly after birth (5-6 days postnatal age). In each animal we measured unanesthetized myocardial perfusion and oxygen delivery using a circumflex artery flow probe. We used inflatable occluders and adenosine to determine coronary conductance and flow reserve. In a subset of animals, we used myocardial contrast echocardiography (MCE, anesthetized) to assess its utility as a tool for studying changes in regional myocardial perfusion in normal development. Separate age-matched animals were instrumented with aortic and coronary sinus sampling catheters to determine myocardial oxygen extraction (unanesthetized). With an average of 17 days of developmental time separating our neonatal and fetal cohorts we found that heart-to-body weight ratio was significantly greater in neonates than fetuses. In resting animals, we found significant decreases in weight-normalized perfusion of, and oxygen delivery to, neonatal relative to fetal myocardium. Similar results were seen when measuring baseline MCE-derived perfusion. Pressure-flow relationship studies revealed lower baseline and maximal coronary conductance in neonates than fetuses, with similar coronary flow reserve between groups. There was greater oxygen extraction in neonates than fetuses. Combined analysis of oxygen extraction with coronary flow suggested greater oxygen consumption by the fetal than neonatal myocardium. We conclude that, during the immediate perinatal period, cardiac growth outpaces coronary microvascular growth resulting in lower capacity for microvascular perfusion in the early neonate.
Collapse
Affiliation(s)
- Matthew W. Hagen
- Center for Developmental HealthOregon Health & Science UniversityPortlandOregonUSA,Knight Cardiovascular Institute, Oregon Health & Science UniversityPortlandOregonUSA
| | - Samantha Louey
- Center for Developmental HealthOregon Health & Science UniversityPortlandOregonUSA,Knight Cardiovascular Institute, Oregon Health & Science UniversityPortlandOregonUSA
| | - Sarah M. Alaniz
- Center for Developmental HealthOregon Health & Science UniversityPortlandOregonUSA
| | - Laura Brown
- Department of PediatricsPerinatal Research CenterUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Jonathan R. Lindner
- Knight Cardiovascular Institute, Oregon Health & Science UniversityPortlandOregonUSA
| | - Sonnet S. Jonker
- Center for Developmental HealthOregon Health & Science UniversityPortlandOregonUSA,Knight Cardiovascular Institute, Oregon Health & Science UniversityPortlandOregonUSA
| |
Collapse
|
27
|
Zhang S, Zhao H, Liu Z, Liu K, Zhu H, Pu W, He L, Wang RA, Zhou B. Monitoring of cell-cell communication and contact history in mammals. Science 2022; 378:eabo5503. [PMID: 36454848 DOI: 10.1126/science.abo5503] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Monitoring of cell-cell communication in multicellular organisms is fundamental to understanding diverse biological processes such as embryogenesis and tumorigenesis. To track cell-cell contacts in vivo, we developed an intercellular genetic technology to monitor cell-cell contact and to trace cell contact histories by permanently marking contacts between cells. In mice, we engineered an artificial Notch ligand into one cell (the sender cell) and an artificial receptor into another cell (the receiver cell). Contact between the sender and receiver cells triggered a synthetic Notch signaling that activated downstream transcriptional programs in the receiver cell, thereby transiently or permanently labeling it. In vivo cell-cell contact was observed during development, tissue homeostasis, and tumor growth. This technology may be useful for studying dynamic in vivo cell-cell contacts and cell fate plasticity.
Collapse
Affiliation(s)
- Shaohua Zhang
- 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
| | - Huan Zhao
- 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
| | - Zixin Liu
- 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
| | - 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 310024, China
| | - Huan Zhu
- 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
| | - Wenjuan Pu
- 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
| | - Lingjuan He
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310030, China
| | - Rong A Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, Division of Vascular Surgery, University of California, San Francisco, CA 94143, USA
| | - 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
- 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 310024, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
28
|
D'Amato G, Phansalkar R, Naftaly JA, Fan X, Amir ZA, Rios Coronado PE, Cowley DO, Quinn KE, Sharma B, Caron KM, Vigilante A, Red-Horse K. Endocardium-to-coronary artery differentiation during heart development and regeneration involves sequential roles of Bmp2 and Cxcl12/Cxcr4. Dev Cell 2022; 57:2517-2532.e6. [PMID: 36347256 PMCID: PMC9833645 DOI: 10.1016/j.devcel.2022.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/28/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
Endocardial cells lining the heart lumen are coronary vessel progenitors during embryogenesis. Re-igniting this developmental process in adults could regenerate blood vessels lost during cardiac injury, but this requires additional knowledge of molecular mechanisms. Here, we use mouse genetics and scRNA-seq to identify regulators of endocardial angiogenesis and precisely assess the role of CXCL12/CXCR4 signaling. Time-specific lineage tracing demonstrated that endocardial cells differentiated into coronary endothelial cells primarily at mid-gestation. A new mouse line reporting CXCR4 activity-along with cell-specific gene deletions-demonstrated it was specifically required for artery morphogenesis rather than angiogenesis. Integrating scRNA-seq data of endocardial-derived coronary vessels from mid- and late-gestation identified a Bmp2-expressing transitioning population specific to mid-gestation. Bmp2 stimulated endocardial angiogenesis in vitro and in injured neonatal mouse hearts. Our data demonstrate how understanding the molecular mechanisms underlying endocardial angiogenesis can identify new potential therapeutic targets promoting revascularization of the injured heart.
Collapse
Affiliation(s)
- Gaetano D'Amato
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Ragini Phansalkar
- Department of Biology, Stanford University, Stanford, CA, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Xiaochen Fan
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Zhainib A Amir
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Dale O Cowley
- Animal Models Core, University of North Carolina, Chapel Hill, NC, USA
| | - Kelsey E Quinn
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bikram Sharma
- Department of Biology, Ball State University, Muncie, IN, USA
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alessandra Vigilante
- Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, King's College London, London, UK
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford, CA, USA.
| |
Collapse
|
29
|
Neffeová K, Olejníčková V, Naňka O, Kolesová H. Development and diseases of the coronary microvasculature and its communication with the myocardium. WIREs Mech Dis 2022; 14:e1560. [DOI: 10.1002/wsbm.1560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 04/12/2022] [Accepted: 04/27/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Kristýna Neffeová
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
| | - Veronika Olejníčková
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
- Institute of Physiology Czech Academy of Science Prague Czech Republic
| | - Ondřej Naňka
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
| | - Hana Kolesová
- Institute of Anatomy, First Faculty of Medicine Charles University Prague Czech Republic
- Institute of Physiology Czech Academy of Science Prague Czech Republic
| |
Collapse
|
30
|
Phansalkar R, Red-Horse K. A new resource for human coronary vessel development. Cardiovasc Res 2022; 118:2875-2876. [PMID: 35726909 DOI: 10.1093/cvr/cvac094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
31
|
Hypoxia promotes a perinatal-like progenitor state in the adult murine epicardium. Sci Rep 2022; 12:9250. [PMID: 35661120 PMCID: PMC9166725 DOI: 10.1038/s41598-022-13107-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/20/2022] [Indexed: 11/22/2022] Open
Abstract
The epicardium is a reservoir of progenitors that give rise to coronary vasculature and stroma during development and mediates cardiac vascular repair. However, its role as a source of progenitors in the adult mammalian heart remains unclear due to lack of clear lineage markers and single-cell culture systems to elucidate epicardial progeny cell fate. We found that in vivo exposure of mice to physiological hypoxia induced adult epicardial cells to re-enter the cell cycle and to express a subset of developmental genes. Multiplex single cell transcriptional profiling revealed a lineage relationship between epicardial cells and smooth muscle, stromal cells, as well as cells with an endothelial-like fate. We found that physiological hypoxia promoted a perinatal-like progenitor state in the adult murine epicardium. In vitro clonal analyses of purified epicardial cells showed that cell growth and subsequent differentiation is dependent upon hypoxia, and that resident epicardial cells retain progenitor identity in the adult mammalian heart with self-renewal and multilineage differentiation potential. These results point to a source of progenitor cells in the adult heart that can be stimulated in vivo and provide an in vitro model for further studies.
Collapse
|
32
|
Xu YH, Feng YF, Zou R, Yuan F, Yuan YZ. Silencing of YAP attenuates pericyte-myofibroblast transition and subretinal fibrosis in experimental model of choroidal neovascularization. Cell Biol Int 2022; 46:1249-1263. [PMID: 35475568 DOI: 10.1002/cbin.11809] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/02/2022] [Indexed: 11/07/2022]
Abstract
Age-related macular degeneration (AMD) is the main reason of irreversible vision loss in the elderly. The subretinal fibrosis subsequent to choroidal neovascularization (CNV) is an important feature in the late stage of wet AMD and is considered to be one reason for incomplete response to anti-VEGF drugs. Recent studies have shown that pericyte-myofibroblast transition (PMT) is an important pathological process involving fibrotic diseases of various organs. However, the specific role and mechanism of PMT in the subretinal fibrosis of CNV have not been clarified. It has been clear that the Hippo pathway along with its downstream effector Yes-associated protein (YAP) plays an important role in both epithelial and endothelial myofibroblast development. Therefore, we speculate whether YAP participates in PMT of pericytes and promotes fibrosis of CNV. In this study, experimental CNV was induced by laser photocoagulation in C57BL/6J (B6) mice, and aberrant YAP overexpression was detected in the retinal pigment epithelial/choroid/sclera tissues of the laser-injured eyes. YAP knockdown reduced the proliferation, migration, and differentiation of human retinal microvascular pericytes in vitro. It also reduced subretinal fibrosis of laser-induced CNV in vivo. Moreover, by proteomics-based analysis of pericyte conditioned medium (PC-CM) and bioinformatic analyses, we identified that the crosstalk between Hippo/YAP and MAPK/Erk was involved in expression of filamin A in hypoxic pericytes. These findings suggest that Hippo/YAP and MAPK/Erk are linked together to mediate pericyte proliferation, migration as well as differentiation, which may embody potential implications for treatment in diseases related to CNV.
Collapse
Affiliation(s)
- Ya-Hui Xu
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Ophthalmology, Northern Jiangsu Peoples' Hospital, Yangzhou, China
| | - Yi-Fan Feng
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rong Zou
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fei Yuan
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan-Zhi Yuan
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Ophthalmology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| |
Collapse
|
33
|
Hulikova A, Park KC, Loonat AA, Gunadasa-Rohling M, Curtis MK, Chung YJ, Wilson A, Carr CA, Trafford AW, Fournier M, Moshnikova A, Andreev OA, Reshetnyak YK, Riley PR, Smart N, Milne TA, Crump NT, Swietach P. Alkaline nucleoplasm facilitates contractile gene expression in the mammalian heart. Basic Res Cardiol 2022; 117:17. [PMID: 35357563 PMCID: PMC8971196 DOI: 10.1007/s00395-022-00924-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 03/04/2022] [Accepted: 03/11/2022] [Indexed: 01/31/2023]
Abstract
Cardiac contractile strength is recognised as being highly pH-sensitive, but less is known about the influence of pH on cardiac gene expression, which may become relevant in response to changes in myocardial metabolism or vascularization during development or disease. We sought evidence for pH-responsive cardiac genes, and a physiological context for this form of transcriptional regulation. pHLIP, a peptide-based reporter of acidity, revealed a non-uniform pH landscape in early-postnatal myocardium, dissipating in later life. pH-responsive differentially expressed genes (pH-DEGs) were identified by transcriptomics of neonatal cardiomyocytes cultured over a range of pH. Enrichment analysis indicated "striated muscle contraction" as a pH-responsive biological process. Label-free proteomics verified fifty-four pH-responsive gene-products, including contractile elements and the adaptor protein CRIP2. Using transcriptional assays, acidity was found to reduce p300/CBP acetylase activity and, its a functional readout, inhibit myocardin, a co-activator of cardiac gene expression. In cultured myocytes, acid-inhibition of p300/CBP reduced H3K27 acetylation, as demonstrated by chromatin immunoprecipitation. H3K27ac levels were more strongly reduced at promoters of acid-downregulated DEGs, implicating an epigenetic mechanism of pH-sensitive gene expression. By tandem cytoplasmic/nuclear pH imaging, the cardiac nucleus was found to exercise a degree of control over its pH through Na+/H+ exchangers at the nuclear envelope. Thus, we describe how extracellular pH signals gain access to the nucleus and regulate the expression of a subset of cardiac genes, notably those coding for contractile proteins and CRIP2. Acting as a proxy of a well-perfused myocardium, alkaline conditions are permissive for expressing genes related to the contractile apparatus.
Collapse
Affiliation(s)
- Alzbeta Hulikova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Kyung Chan Park
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Aminah A Loonat
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Mala Gunadasa-Rohling
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - M Kate Curtis
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Yu Jin Chung
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Abigail Wilson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Andrew W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Marjorie Fournier
- Department of Biochemistry, Advanced Proteomics Facility, University of Oxford, Oxford, UK
| | - Anna Moshnikova
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Oleg A Andreev
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Yana K Reshetnyak
- Physics Department, University of Rhode Island, 2 Lippitt Rd, Kingston, RI, 02881, USA
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK
| | - Thomas A Milne
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford, UK
| | - Nicholas T Crump
- MRC Molecular Haematology Unit, Radcliffe Department of Medicine, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, University of Oxford, Oxford, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
34
|
Tian X, Zhou B. Coronary vessel formation in development and regeneration: origins and mechanisms. J Mol Cell Cardiol 2022; 167:67-82. [DOI: 10.1016/j.yjmcc.2022.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/12/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
35
|
McCracken IR, Dobie R, Bennett M, Passi R, Beqqali A, Henderson NC, Mountford JC, Riley PR, Ponting CP, Smart N, Brittan M, Baker AH. Mapping the developing human cardiac endothelium at single-cell resolution identifies MECOM as a regulator of arteriovenous gene expression. Cardiovasc Res 2022; 118:2960-2972. [PMID: 35212715 PMCID: PMC9648824 DOI: 10.1093/cvr/cvac023] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/24/2022] [Indexed: 11/25/2022] Open
Abstract
AIMS Coronary vasculature formation is a critical event during cardiac development, essential for heart function throughout perinatal and adult life. However, current understanding of coronary vascular development has largely been derived from transgenic mouse models. The aim of this study was to characterize the transcriptome of the human foetal cardiac endothelium using single-cell RNA sequencing (scRNA-seq) to provide critical new insights into the cellular heterogeneity and transcriptional dynamics that underpin endothelial specification within the vasculature of the developing heart. METHODS AND RESULTS We acquired scRNA-seq data of over 10 000 foetal cardiac endothelial cells (ECs), revealing divergent EC subtypes including endocardial, capillary, venous, arterial, and lymphatic populations. Gene regulatory network analyses predicted roles for SMAD1 and MECOM in determining the identity of capillary and arterial populations, respectively. Trajectory inference analysis suggested an endocardial contribution to the coronary vasculature and subsequent arterialization of capillary endothelium accompanied by increasing MECOM expression. Comparative analysis of equivalent data from murine cardiac development demonstrated that transcriptional signatures defining endothelial subpopulations are largely conserved between human and mouse. Comprehensive characterization of the transcriptional response to MECOM knockdown in human embryonic stem cell-derived EC (hESC-EC) demonstrated an increase in the expression of non-arterial markers, including those enriched in venous EC. CONCLUSIONS scRNA-seq of the human foetal cardiac endothelium identified distinct EC populations. A predicted endocardial contribution to the developing coronary vasculature was identified, as well as subsequent arterial specification of capillary EC. Loss of MECOM in hESC-EC increased expression of non-arterial markers, suggesting a role in maintaining arterial EC identity.
Collapse
Affiliation(s)
- Ian R McCracken
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK,Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Ross Dobie
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Matthew Bennett
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Rainha Passi
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Abdelaziz Beqqali
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK,MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | | | - Paul R Riley
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Chris P Ponting
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Nicola Smart
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Mairi Brittan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | |
Collapse
|
36
|
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: 12] [Impact Index Per Article: 4.0] [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
|
37
|
Hou S, Li Z, Dong J, Gao Y, Chang Z, Ding X, Li S, Li Y, Zeng Y, Xin Q, Wang B, Ni Y, Ning X, Hu Y, Fan X, Hou Y, Li X, Wen L, Zhou B, Liu B, Tang F, Lan Y. Heterogeneity in endothelial cells and widespread venous arterialization during early vascular development in mammals. Cell Res 2022; 32:333-348. [PMID: 35079138 PMCID: PMC8975889 DOI: 10.1038/s41422-022-00615-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/23/2021] [Indexed: 12/11/2022] Open
Abstract
AbstractArteriogenesis rather than unspecialized capillary expansion is critical for restoring effective circulation to compromised tissues in patients. Deciphering the origin and specification of arterial endothelial cells during embryonic development will shed light on the understanding of adult arteriogenesis. However, during early embryonic angiogenesis, the process of endothelial diversification and molecular events underlying arteriovenous fate settling remain largely unresolved in mammals. Here, we constructed the single-cell transcriptomic landscape of vascular endothelial cells (VECs) during the time window for the occurrence of key vasculogenic and angiogenic events in both mouse and human embryos. We uncovered two distinct arterial VEC types, the major artery VECs and arterial plexus VECs, and unexpectedly divergent arteriovenous characteristics among VECs that are located in morphologically undistinguishable vascular plexus intra-embryonically. Using computational prediction and further lineage tracing of venous-featured VECs with a newly developed Nr2f2CrexER mouse model and a dual recombinase-mediated intersectional genetic approach, we revealed early and widespread arterialization from the capillaries with considerable venous characteristics. Altogether, our findings provide unprecedented and comprehensive details of endothelial heterogeneity and lineage relationships at early angiogenesis stages, and establish a new model regarding the arteriogenesis behaviors of early intra-embryonic vasculatures.
Collapse
|
38
|
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
|
39
|
Chowdhury K, Lai SL, Marín-Juez R. Modulation of VEGFA Signaling During Heart Regeneration in Zebrafish. Methods Mol Biol 2022; 2475:297-312. [PMID: 35451767 DOI: 10.1007/978-1-0716-2217-9_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Over the last decades, myocardial infarction and heart failure have accounted every year for millions of deaths worldwide. After a coronary occlusion, the lack of blood supply to downstream muscle leads to cell death and scarring. To date, several pro-angiogenic factors have been tested to stimulate reperfusion of the affected myocardium, VEGFA being one of the most extensively studied. Given the unsuccessful outcomes of clinical trials, understanding how cardiac revascularization takes place in models with endogenous regenerative capacity holds the key to devising more efficient therapies. Here, we summarize the main findings on VEGFA's role during cardiac repair and regeneration, with a particular focus on zebrafish as a regenerative model. Moreover, we provide a comprehensive overview of available tools to modulate Vegfa expression and action in zebrafish regeneration studies. Understanding the role of Vegfa during zebrafish heart regeneration may help devise efficient therapies and circumvent current limitations in using VEGFA for therapeutic angiogenesis approaches.
Collapse
Affiliation(s)
- Kaushik Chowdhury
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Shih-Lei Lai
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Rubén Marín-Juez
- Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montreal, QC, Canada.
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
40
|
Phansalkar R, Krieger J, Zhao M, Kolluru SS, Jones RC, Quake SR, Weissman I, Bernstein D, Winn VD, D'Amato G, Red-Horse K. Coronary blood vessels from distinct origins converge to equivalent states during mouse and human development. eLife 2021; 10:e70246. [PMID: 34910626 PMCID: PMC8673841 DOI: 10.7554/elife.70246] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 12/02/2021] [Indexed: 12/17/2022] Open
Abstract
Most cell fate trajectories during development follow a diverging, tree-like branching pattern, but the opposite can occur when distinct progenitors contribute to the same cell type. During this convergent differentiation, it is unknown if cells 'remember' their origins transcriptionally or whether this influences cell behavior. Most coronary blood vessels of the heart develop from two different progenitor sources-the endocardium (Endo) and sinus venosus (SV)-but whether transcriptional or functional differences related to origin are retained is unknown. We addressed this by combining lineage tracing with single-cell RNA sequencing (scRNAseq) in embryonic and adult mouse hearts. Shortly after coronary development begins, capillary endothelial cells (ECs) transcriptionally segregated into two states that retained progenitor-specific gene expression. Later in development, when the coronary vasculature is well established but still remodeling, capillary ECs again segregated into two populations, but transcriptional differences were primarily related to tissue localization rather than lineage. Specifically, ECs in the heart septum expressed genes indicative of increased local hypoxia and decreased blood flow. Adult capillary ECs were more homogeneous with respect to both lineage and location. In agreement, SV- and Endo-derived ECs in adult hearts displayed similar responses to injury. Finally, scRNAseq of developing human coronary vessels indicated that the human heart followed similar principles. Thus, over the course of development, transcriptional heterogeneity in coronary ECs is first influenced by lineage, then by location, until heterogeneity declines in the homeostatic adult heart. These results highlight the plasticity of ECs during development, and the validity of the mouse as a model for human coronary development.
Collapse
Affiliation(s)
- Ragini Phansalkar
- Department of Genetics, Stanford University School of MedicineStanfordUnited States
- Department of Biology, Stanford UniversityStanfordUnited States
| | | | - Mingming Zhao
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| | - Sai Saroja Kolluru
- Department of Bioengineering and Department of Applied Physics, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering and Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering and Department of Applied Physics, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubStanfordUnited States
| | - Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| | - Daniel Bernstein
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of MedicineStanfordUnited States
| | - Gaetano D'Amato
- Department of Biology, Stanford UniversityStanfordUnited States
| | - Kristy Red-Horse
- Department of Biology, Stanford UniversityStanfordUnited States
- Stanford Cardiovascular Institute, Stanford University School of MedicineStanfordUnited States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
41
|
Developmental angiocrine diversification of endothelial cells for organotypic regeneration. Dev Cell 2021; 56:3042-3051. [PMID: 34813766 DOI: 10.1016/j.devcel.2021.10.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023]
Abstract
Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.
Collapse
|
42
|
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
|
43
|
Histamine Deficiency Promotes Myofibroblasts Transformation from HDC-Expressing CD11b + Myeloid Cells in Injured Hearts Post Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:621-634. [PMID: 34734351 DOI: 10.1007/s12265-021-10172-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/08/2021] [Indexed: 10/19/2022]
Abstract
Myocardial infarction (MI) is a significant contributor to the development of heart failure. Histidine decarboxylase (HDC), the unique enzyme that converts L-histidine to histamine, is highly expressed in CD11b+ immature myeloid cells. However, the relationship between HDC-expressing macrophages and cardiac myofibroblasts remains to be explained. Here, we demonstrate that the GFP (green fluorescent protein)-labeled HDC+CD11b+ myeloid precursors and their descendants could differentiate into fibroblast-like cells in myocardial interstitium. Furthermore, we prove that CD11b+Ly6C+ monocytes/macrophages, but not CD11b+Ly6G+ granulocytes, are identified as the main cellular source for bone marrow-derived myofibroblast transformation, which could be regulated via histamine H1 and H2 receptor-dependent signaling pathways. Using HDC knockout mice, we find that histamine deficiency promotes myofibroblast transformation from Ly6C+ macrophages and cardiac fibrosis partly through upregulating the expression of Krüppel-like factor 5 (KLF5). Taken together, our data uncover a central role of HDC in regulating bone marrow-derived macrophage-to-myofibroblast transformation but also identify a histamine receptor (HR)-KLF5 related signaling pathway that mediates myocardial fibrosis post-MI. CD11b+Ly6C+ monocytes/macrophages are the main cellular source for bone marrow-derived myofibroblast transformation. Histamine inhibits myofibroblasts transformation via H1R and H2R-dependent signaling pathways, and ameliorates cardiac fibrosis partly through upregulating KLF5 expression.
Collapse
|
44
|
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
|
45
|
Lu P, Wang Y, Liu Y, Wang Y, Wu B, Zheng D, Harvey RP, Zhou B. Perinatal angiogenesis from pre-existing coronary vessels via DLL4-NOTCH1 signalling. Nat Cell Biol 2021; 23:967-977. [PMID: 34497373 DOI: 10.1038/s41556-021-00747-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 07/27/2021] [Indexed: 12/13/2022]
Abstract
New coronary vessels are added to the heart around birth to support postnatal cardiac growth. Here we show that, in late fetal development, the embryonic coronary plexus at the inner myocardium of the ventricles expresses the angiogenic signalling factors VEGFR3 and DLL4 and generates new coronary vessels in neonates. Contrary to a previous model in which the formation of new coronary vessels in neonates from ventricular endocardial cells was proposed, we find that late fetal and neonatal ventricular endocardial cells lack angiogenic potential and do not contribute to new coronary vessels. Instead, we show using lineage-tracing as well as gain- and loss-of-function experiments that the pre-existing embryonic coronary plexus at the inner myocardium undergoes angiogenic expansion through the DLL4-NOTCH1 signalling pathway to vascularize the expanding myocardium. We also show that the pre-existing coronary plexus revascularizes the regenerating neonatal heart through a similar mechanism. These findings provide a different model of neonatal coronary angiogenesis and regeneration, potentially informing cardiovascular medicine.
Collapse
Affiliation(s)
- Pengfei Lu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Yidong Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Yifeng Wang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, New York, NY, USA
| | - Richard P Harvey
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
- St Vincent's Clinical School, and School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA.
- Departments of Pediatrics (Pediatric Genetic Medicine) and Medicine (Cardiology), Albert Einstein College of Medicine, New York, NY, USA.
- Wilf Family Cardiovascular Research Institute and Einstein Institute for Aging Research, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
46
|
Nawrocki MJ, Jopek K, Zdun M, Mozdziak P, Jemielity M, Perek B, Bukowska D, Kempisty B. Expression Profile of Genes Encoding Proteins Involved in Regulation of Vasculature Development and Heart Muscle Morphogenesis-A Transcriptomic Approach Based on a Porcine Model. Int J Mol Sci 2021; 22:ijms22168794. [PMID: 34445494 PMCID: PMC8395751 DOI: 10.3390/ijms22168794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022] Open
Abstract
Despite significant advances in treatment of acute coronary syndromes (ACS) many subjects still develop heart failure due to significantly reduced ejection fraction. Currently, there are no commonly available treatment strategies that replace the infarcted/dysfunctional myocardium. Therefore, understanding the mechanisms that control the regeneration of the heart muscle is important. The development of new coronary vessels plays a pivotal role in cardiac regeneration. Employing microarray expression assays and RT-qPCR validation expression pattern of genes in long-term primary cultured cells isolated form the right atrial appendage (RAA) and right atrium (RA) was evaluated. After using DAVID software, it indicated the analysis expression profiles of genes involved in ontological groups such as: “angiogenesis”, “blood vessel morphogenesis”, “circulatory system development”, “regulation of vasculature development”, and “vasculature development” associated with the process of creation new blood vessels. The performed transcriptomic comparative analysis between two different compartments of the heart muscle allowed us to indicate the presence of differences in the expression of key transcripts depending on the cell source. Increases in culture intervals significantly increased expression of SFRP2, PRRX1 genes and some other genes involved in inflammatory process, such as: CCL2, IL6, and ROBO1. Moreover, the right atrial appendage gene encoding lysyl oxidase (LOX) showed much higher expression compared to the pre-cultivation state.
Collapse
Affiliation(s)
- Mariusz J. Nawrocki
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland;
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland;
| | - Maciej Zdun
- Department of Basic and Preclinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland;
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA;
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Marek Jemielity
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-848 Poznań, Poland; (M.J.); (B.P.)
| | - Bartłomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, 61-848 Poznań, Poland; (M.J.); (B.P.)
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland;
| | - Bartosz Kempisty
- Department of Anatomy, Poznan University of Medical Sciences, 60-781 Poznań, Poland;
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznań, Poland;
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Toruń, Poland
- Correspondence: ; Tel.: +48-61-8546418; Fax: +48-61-8546440
| |
Collapse
|
47
|
Kim H, Wang M, Paik DT. Endothelial-Myocardial Angiocrine Signaling in Heart Development. Front Cell Dev Biol 2021; 9:697130. [PMID: 34277641 PMCID: PMC8281241 DOI: 10.3389/fcell.2021.697130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/10/2021] [Indexed: 12/23/2022] Open
Abstract
Vascular endothelial cells are a multifunctional cell type with organotypic specificity in their function and structure. In this review, we discuss various subpopulations of endothelial cells in the mammalian heart, which spatiotemporally regulate critical cellular and molecular processes of heart development via unique sets of angiocrine signaling pathways. In particular, elucidation of intercellular communication among the functional cell types in the developing heart has recently been accelerated by the use of single-cell sequencing. Specifically, we overview the heterogeneic nature of cardiac endothelial cells and their contribution to heart tube and chamber formation, myocardial trabeculation and compaction, and endocardial cushion and valve formation via angiocrine pathways.
Collapse
Affiliation(s)
- Hyeonyu Kim
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Mingqiang Wang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
48
|
Stone OA, Zhou B, Red-Horse K, Stainier DYR. Endothelial ontogeny and the establishment of vascular heterogeneity. Bioessays 2021; 43:e2100036. [PMID: 34145927 DOI: 10.1002/bies.202100036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
The establishment of distinct cellular identities was pivotal during the evolution of Metazoa, enabling the emergence of an array of specialized tissues with different functions. In most animals including vertebrates, cell specialization occurs in response to a combination of intrinsic (e.g., cellular ontogeny) and extrinsic (e.g., local environment) factors that drive the acquisition of unique characteristics at the single-cell level. The first functional organ system to form in vertebrates is the cardiovascular system, which is lined by a network of endothelial cells whose organ-specific characteristics have long been recognized. Recent genetic analyses at the single-cell level have revealed that heterogeneity exists not only at the organ level but also between neighboring endothelial cells. Thus, how endothelial heterogeneity is established has become a key question in vascular biology. Drawing upon evidence from multiple organ systems, here we will discuss the role that lineage history may play in establishing endothelial heterogeneity.
Collapse
Affiliation(s)
- Oliver A Stone
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Bin Zhou
- The State Key Laboratory of Cell Biology, CAS Center for Excellence on Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kristy Red-Horse
- Department of Biology, Stanford Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
49
|
Feng F, Feng X, Zhang D, Li Q, Yao L. Matrix Stiffness Induces Pericyte-Fibroblast Transition Through YAP Activation. Front Pharmacol 2021; 12:698275. [PMID: 34135765 PMCID: PMC8202079 DOI: 10.3389/fphar.2021.698275] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular pericytes, important mural cells that retain progenitor cell properties and protect vascular integrity in healthy tissues, are often associated with tumor development, but their functions in cancer invasion remain elusive. One prominent outcome of tumor occurrence is that the microenvironment of the lesion often stiffens, which could change resident cell behavior. Here, we found pericytes are matrix stiffness-responsive and mechanical stimuli induce pericyte-fibroblast transition (PFT). Soft PA gels that mimic the stiffness of healthy tissues retain the identity and behavior of pericytes, whereas stiff PA gels that reflect the stiffness of tumorous tissues promote PFT and the mobility and invasiveness of the cells. Matrix stiffness-induced PFT depends on the activation of YAP (Yes-associated protein), a transcription factor, which, upon receiving mechanical signals, transfers from cytoplasm to nucleus to mediate cell transcriptional activities. Our result reveals a mechanism through which vascular pericytes convert to fibroblasts and migrate away from vasculatures to help tumor development, and thus targeting matrix stiffness-induced PFT may offer a new perspective to the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Feng Feng
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xueyan Feng
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Di Zhang
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qilong Li
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Li Yao
- State Key Laboratory for Structural Chemistry of Unstable and Stable Species, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China.,School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
50
|
Guo P, Liu Y, Xu X, Ma G, Hou X, Fan Y, Zhang M. Coronary hypercontractility to acidosis owes to the greater activity of TMEM16A/ANO1 in the arterial smooth muscle cells. Biomed Pharmacother 2021; 139:111615. [PMID: 34243598 DOI: 10.1016/j.biopha.2021.111615] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/05/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Severe acidosis deteriorates cardiac injury. Rat coronary arteries (RCAs) are unusually hypercontractive to extracellular (o) acidosis (EA). TMEM16A-encoded anoctamin 1 (ANO1), a Ca2+-activated chloride channel (CaCC), plays an important role in regulating coronary arterial tension. PURPOSE We tested the possibility that the activation of CaCCs in the arterial smooth muscle cell (ASMC) contributes to EA-induced RCA constriction. METHODS ANO1 expression was detected with immunofluorescence staining and Western blot. TMEM16A mRNA was assessed with quantitative Real-Time PCR. Cl- currents and membrane potentials were quantified with a patch clamp. The vascular tension was recorded with a myograph. Intracellular (i) level of Cl- and Ca2+ was measured with fluorescent molecular probes. RESULTS ANO1 was expressed in all tested arterial myocytes, but was much more abundant in RCA ASMCs as compared with ASMCs isolated from rat cerebral basilar, intrarenal and mesenteric arteries. EA reduced [Cl-]i levels, augmented CaCC currents exclusively in RCA ASMCs and depolarized RCA ASMCs to a greater extent. Cl- deprivation, which depleted [Cl-]i by incubating the arteries or their ASMCs in Cl--free bath solution, decreased EA-induced [Cl-]i reduction, diminished EA-induced CaCC augmentation and time-dependently depressed EA-induced RCA constriction. Inhibitor studies showed that these EA-induced effects including RCA constriction, CaCC current augmentation, [Cl-]i reduction and/or [Ca2+]i elevation were depressed by various Cl- channel blockers, [Ca2+]i release inhibitors and L-type voltage-gated Ca2+ channel inhibitor nifedipine. ANO1 antibody attenuated all observed changes induced by EA in RCA ASMCs. CONCLUSION The greater activity of RCA ASMC CaCCs complicated with an enhanced Ca2+ mobilization from both [Ca2+]i release and [Ca2+]o influx plays a pivotal role in the distinctive hypercontractility of RCAs to acidosis. Translation of these findings to human beings may lead to a new conception in our understanding and treating cardiac complications in severe acidosis.
Collapse
Affiliation(s)
- Pengmei Guo
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China
| | - Yu Liu
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China
| | - Xiaojia Xu
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China
| | - Guijin Ma
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China
| | - Xiaomin Hou
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China
| | - Yanying Fan
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China.
| | - Mingsheng Zhang
- Department of Pharmacology, Shanxi Medical University, Xinjiannanlu 56, Taiyuan 030001, Shanxi Province, China.
| |
Collapse
|