1
|
Arduini A, Fleming SJ, Xiao L, Hall AW, Akkad AD, Chaffin MD, Bendinelli KJ, Tucker NR, Papangeli I, Mantineo H, Flores-Bringas P, Babadi M, Stegmann CM, García-Cardeña G, Lindsay ME, Klattenhoff C, Ellinor PT. Transcriptional profile of the rat cardiovascular system at single-cell resolution. Cell Rep 2025; 44:115091. [PMID: 39709602 PMCID: PMC11781962 DOI: 10.1016/j.celrep.2024.115091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/24/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024] Open
Abstract
We sought to characterize cellular composition across the cardiovascular system of the healthy Wistar rat, an important model in preclinical cardiovascular research. We performed single-nucleus RNA sequencing (snRNA-seq) in 78 samples in 10 distinct regions, including the four chambers of the heart, ventricular septum, sinoatrial node, atrioventricular node, aorta, pulmonary artery, and pulmonary veins, which produced 505,835 nuclei. We identified 26 distinct cell types and additional subtypes, with different cellular composition across cardiac regions and tissue-specific transcription for each cell type. Several cell subtypes were region specific, including a subtype of vascular smooth muscle cells enriched in the large vasculature. We observed tissue-enriched cellular communication networks, including heightened Nppa-Npr1/2/3 signaling in the sinoatrial node. The existence of tissue-restricted cell types suggests regional regulation of cardiovascular physiology. Our detailed transcriptional characterization of each cell type offers the potential to identify novel therapeutic targets and improve preclinical models of cardiovascular disease.
Collapse
Affiliation(s)
- Alessandro Arduini
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA
| | - Stephen J Fleming
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ling Xiao
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amelia W Hall
- Gene Regulation Observatory, The Broad Institute, Cambridge, MA 02142, USA
| | - Amer-Denis Akkad
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA 02142, USA
| | - Mark D Chaffin
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA
| | - Kayla J Bendinelli
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA
| | | | - Irinna Papangeli
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA 02142, USA
| | - Helene Mantineo
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | - Mehrtash Babadi
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Guillermo García-Cardeña
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02215, USA
| | - Mark E Lindsay
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Carla Klattenhoff
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA 02142, USA
| | - Patrick T Ellinor
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Cardiology Division, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
2
|
Zhang Z, Ma X, La Y, Guo X, Chu M, Bao P, Yan P, Wu X, Liang C. Advancements in the Application of scRNA-Seq in Breast Research: A Review. Int J Mol Sci 2024; 25:13706. [PMID: 39769466 PMCID: PMC11677372 DOI: 10.3390/ijms252413706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/10/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Single-cell sequencing technology provides apparent advantages in cell population heterogeneity, allowing individuals to better comprehend tissues and organs. Sequencing technology is currently moving beyond the standard transcriptome to the single-cell level, which is likely to bring new insights into the function of breast cells. In this study, we examine the primary cell types involved in breast development, as well as achievements in the study of scRNA-seq in the microenvironment, stressing the finding of novel cell subsets using single-cell approaches and analyzing the problems and solutions to scRNA-seq. Furthermore, we are excited about the field's promising future.
Collapse
Affiliation(s)
- Zhenyu Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xiaoming Ma
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Yongfu La
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xian Guo
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Min Chu
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Pengjia Bao
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Ping Yan
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Xiaoyun Wu
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| | - Chunnian Liang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China;
- Key Laboratory for Yak Genetics, Breeding, and Reproduction Engineering of Gansu Province, Gansu Provincial Key Laboratory of Yak Breeding Engineering, Lanzhou Institute of Animal Husbandry and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Lanzhou 730070, China; (X.M.); (Y.L.); (X.G.); (M.C.); (P.B.); (P.Y.); (X.W.)
- Key Laboratory of Animal Genetics and Breeding on Tibetan Plateau, Ministry of Agriculture and Rural Affairs, Lanzhou 730070, China
| |
Collapse
|
3
|
Brückner A, Brandtner A, Rieck S, Matthey M, Geisen C, Fels B, Stei M, Kusche-Vihrog K, Fleischmann BK, Wenzel D. Site-specific genetic and functional signatures of aortic endothelial cells at aneurysm predilection sites in healthy and AngII ApoE -/- mice. Angiogenesis 2024; 27:719-738. [PMID: 38965173 PMCID: PMC11564227 DOI: 10.1007/s10456-024-09933-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 06/16/2024] [Indexed: 07/06/2024]
Abstract
Aortic aneurysm is characterized by a pathological dilation at specific predilection sites of the vessel and potentially results in life-threatening vascular rupture. Herein, we established a modified "Häutchen method" for the local isolation of endothelial cells (ECs) from mouse aorta to analyze their spatial heterogeneity and potential role in site-specific disease development. When we compared ECs from aneurysm predilection sites of healthy mice with adjacent control segments we found regulation of genes related to extracellular matrix remodeling, angiogenesis and inflammation, all pathways playing a critical role in aneurysm development. We also detected enhanced cortical stiffness of the endothelium at these sites. Gene expression of ECs from aneurysms of the AngII ApoE-/- model when compared to sham animals mimicked expression patterns from predilection sites of healthy animals. Thus, this work highlights a striking genetic and functional regional heterogeneity in aortic ECs of healthy mice, which defines the location of aortic aneurysm formation in disease.
Collapse
Affiliation(s)
- Alexander Brückner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Adrian Brandtner
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Sarah Rieck
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany
| | - Caroline Geisen
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Benedikt Fels
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Marta Stei
- Heart Center Bonn, Clinic for Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Kristina Kusche-Vihrog
- Institute of Physiology, University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Partner SiteHamburg/Luebeck/Kiel, Luebeck, Germany
| | - Bernd K Fleischmann
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany
| | - Daniela Wenzel
- Life&Brain Center, Medical Faculty, Institute of Physiology I, University of Bonn, Bonn, Germany.
- Department of Systems Physiology, Medical Faculty, Institute of Physiology, Ruhr University of Bochum, Universitätsstr. 150, 44801, Bochum, Germany.
| |
Collapse
|
4
|
Mao A, Li Z, Shi X, Zhang K, Kan H, Geng L, He D. Complement Factor C1q Mediates Vascular Endothelial Dysfunction in STZ-Induced Diabetic Mice. Diabetes 2024; 73:1527-1536. [PMID: 38869460 DOI: 10.2337/db23-0981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
Diabetes is a significant global public health issue with implications for vascular endothelial cells (ECs) dysfunction and the subsequent development and advancement of diabetes complications. This study aims to compare the cellular and molecular properties of the aorta in normal and streptozotocin (STZ)-induced diabetic mice, with a focus on elucidating potential mechanism underlying EC dysfunction. Here, we performed a single-cell RNA sequencing survey of 32,573 cells from the aorta of normal and STZ-induced diabetic mice. We found a compendium of 10 distinct cell types, mainly ECs, smooth muscle cells, fibroblast, pericyte, immune cells, and stromal cells. As the diabetes condition progressed, we observed a subpopulation of aortic ECs that exhibited significantly elevated expression of complement (C) molecule C1qa compared with their healthy counterparts. This increased expression of C1qa was found to induce reactive oxygen species (ROS) production, facilitate EC migration and increased permeability, and impair the vasodilation within the aortic segment of mice. Furthermore, AAV-Tie2-shRNA-C1qa was administered into diabetic mice by tail vein injection, showing that inhibition of C1qa in the endothelium led to a reduction in ROS production, decreased vascular permeability, and improved vasodilation. Collectively, these findings highlight the crucial involvement of C1qa in endothelial dysfunction associated with diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zicheng Li
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xiaoming Shi
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongxu He
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
5
|
Koleilat I, Dalmia V, Batarseh P, Rai A, Carnevale M, Phair J, Indes J. Large-Diameter Fenestrated Endograft Repair of Abdominal Aortic Aneurysms Is Not Associated With Medium-Term Outcomes. J Surg Res 2024; 296:516-522. [PMID: 38330677 DOI: 10.1016/j.jss.2024.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/11/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Recent data suggests that infrarenal abdominal aortic aneurysm (AAA) endovascular repair (EVAR) with large diameter grafts (LGs) may have a higher risk of endoleak and reintervention. However, this has not been studied extensively for fenestrated endovascular aneurysm repair (fEVAR). We, therefore, sought to evaluate the outcomes of patients undergoing fEVAR with large-diameter endografts. METHODS Patients from the national Vascular Quality Initiative registry who underwent fEVAR for intact juxtarenal AAA were identified. Patients with genetic causes for aneurysms, those with prior aortic surgery, and those undergoing repair for symptomatic or ruptured aneurysms were excluded. Rates of endoleaks and reintervention at periprocedural and long-term follow-up timepoints (9-22 mo) were analyzed in grafts 32 mm or larger (LG) and were compared to those smaller than 32 mm (small diameter graft). RESULTS A total of 693 patients (22.8% LG) were identified. Overall, demographic variables were comparable except LG exhibited a more frequent history of coronary artery disease (32.9% versus 25.4%, P = 0.037). There were no significant differences in the rates of endoleak at procedural completion. Overall survival at 5 y was no different. The rate of reintervention at 1 y was also no different (log-rank P = 0.86). CONCLUSIONS While graft size appears to have an association with outcomes in infrarenal aneurysm repair, the same does not appear to be true for fEVAR. Further studies should evaluate the long-term outcomes associated with LG which could alter the approach to repair of AAA with large neck diameters traditionally treated with standard infrarenal EVAR.
Collapse
Affiliation(s)
- Issam Koleilat
- Department of Surgery, Community Medical Center, RWJ/Barnabas Health, Toms River, New Jersey.
| | - Varun Dalmia
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Paola Batarseh
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Anvit Rai
- Division of Vascular and Endovascular Surgery, Department of Cardiothoracic and Vascular Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| | - Matthew Carnevale
- Division of Vascular and Endovascular Surgery, Department of Cardiothoracic and Vascular Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| | - John Phair
- Division of Vascular Surgery, Department of Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jeff Indes
- Division of Vascular and Endovascular Surgery, Department of Cardiothoracic and Vascular Surgery, Montefiore Medical Center/Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
6
|
Gibson Hughes TA, Dona MSI, Sobey CG, Pinto AR, Drummond GR, Vinh A, Jelinic M. Aortic Cellular Heterogeneity in Health and Disease: Novel Insights Into Aortic Diseases From Single-Cell RNA Transcriptomic Data Sets. Hypertension 2024; 81:738-751. [PMID: 38318714 DOI: 10.1161/hypertensionaha.123.20597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Aortic diseases such as atherosclerosis, aortic aneurysms, and aortic stiffening are significant complications that can have significant impact on end-stage cardiovascular disease. With limited pharmacological therapeutic strategies that target the structural changes in the aorta, surgical intervention remains the only option for some patients with these diseases. Although there have been significant contributions to our understanding of the cellular architecture of the diseased aorta, particularly in the context of atherosclerosis, furthering our insight into the cellular drivers of disease is required. The major cell types of the aorta are well defined; however, the advent of single-cell RNA sequencing provides unrivaled insights into the cellular heterogeneity of each aortic cell type and the inferred biological processes associated with each cell in health and disease. This review discusses previous concepts that have now been enhanced with recent advances made by single-cell RNA sequencing with a focus on aortic cellular heterogeneity.
Collapse
Affiliation(s)
- Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Malathi S I Dona
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Alexander R Pinto
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
- Baker Heart and Diabetes Research Institute, Melbourne, Victoria, Australia (M.S.I.D., A.R.P.)
| | - Grant R Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Antony Vinh
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| | - Maria Jelinic
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC, Australia (T.A.G.H., C.G.S., A.R.P., G.R.D., A.V., M.J.)
| |
Collapse
|
7
|
Ma S, Xie X, Yuan R, Xin Q, Miao Y, Leng SX, Chen K, Cong W. Vascular Aging and Atherosclerosis: A Perspective on Aging. Aging Dis 2024; 16:AD.2024.0201-1. [PMID: 38502584 PMCID: PMC11745439 DOI: 10.14336/ad.2024.0201-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/01/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular aging (VA) is recognized as a pivotal factor in the development and progression of atherosclerosis (AS). Although various epidemiological and clinical research has demonstrated an intimate connection between aging and AS, the candidate mechanisms still require thorough examination. This review adopts an aging-centric perspective to deepen the comprehension of the intricate relationship between biological aging, vascular cell senescence, and AS. Various aging-related physiological factors influence the physical system's reactions, including oxygen radicals, inflammation, lipids, angiotensin II, mechanical forces, glucose levels, and insulin resistance. These factors cause endothelial dysfunction, barrier damage, sclerosis, and inflammation for VA and promote AS via distinct or shared pathways. Furthermore, the increase of senescent cells inside the vascular tissues, caused by genetic damage, dysregulation, secretome changes, and epigenetic modifications, might be the primary cause of VA.
Collapse
Affiliation(s)
- Shudong Ma
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xuena Xie
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Sean Xiao Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA.
| | - Keji Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Weihong Cong
- School of Pharmacy, Macau University of Science and Technology, Macau, China.
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
8
|
Kudryavtseva O, Lyngsø KS, Jensen BL, Dimke H. Nitric oxide, endothelium-derived hyperpolarizing factor, and smooth muscle-dependent mechanisms contribute to magnesium-dependent vascular relaxation in mouse arteries. Acta Physiol (Oxf) 2024; 240:e14096. [PMID: 38258597 DOI: 10.1111/apha.14096] [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: 06/08/2023] [Revised: 09/27/2023] [Accepted: 01/01/2024] [Indexed: 01/24/2024]
Abstract
AIM Magnesium (Mg2+ ) is a vasorelaxant. The underlying physiological mechanisms driving this vasorelaxation remain unclear. Studies were designed to test the hypothesis that multiple signaling pathways including nitric oxide (NO) and endothelium-derived hyperpolarizing factor (EDHF) in endothelial cells as well as Ca2+ antagonization and TRPM7 channels in vascular smooth muscle cells mediate Mg2+ -dependent vessel relaxation. METHODS To uncover these mechanisms, force development was measured ex vivo in aorta rings from mice using isometric wire myography. Concentration responses to Mg2+ were studied in intact and endothelium-denuded aortas. Key findings were confirmed in second-order mesenteric resistance arteries perfused ex vivo using pressure myography. Effects of Mg2+ on NO formation were measured in Chinese Hamster Ovary (CHO) cells, isolated mesenteric vessels, and mouse urine. RESULTS Mg2+ caused a significant concentration-dependent relaxation of aorta rings. This relaxation was attenuated significantly in endothelium-denuded aortas. The endothelium-dependent portion was inhibited by NO and cGMP blockade but not by cyclooxygenase inhibition. Mg2+ stimulated local NO formation in CHO cells and isolated mesenteric vessels without changing urinary NOx levels. High extracellular Mg2+ augmented acetylcholine-induced relaxation. SKCa and IKCa channel blockers apamin and TRAM34 inhibited Mg2+ -dependent relaxation. The endothelium-independent relaxation in aorta rings was inhibited by high extracellular Ca2+ . Combined blockade of NO, SKCa , and IKCa channels significantly reduced Mg2+ -dependent dilatation in mesenteric resistance vessels. CONCLUSIONS In mouse conductance and resistance arteries Mg2+ -induced relaxation is contributed by endothelial NO formation, EDHF pathways, antagonism of Ca2+ in smooth muscle cells, and additional unidentified mechanisms.
Collapse
Affiliation(s)
- Olga Kudryavtseva
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Kristina S Lyngsø
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Boye L Jensen
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
| | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense C, Denmark
- Department of Nephrology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
9
|
Mao A, Zhang K, Kan H, Gao M, Wang Z, Zhou T, Shao J, He D. Single-Cell RNA-Seq Reveals Coronary Heterogeneity and Identifies CD133 +TRPV4 high Endothelial Subpopulation in Regulating Flow-Induced Vascular Tone in Mice. Arterioscler Thromb Vasc Biol 2024; 44:653-665. [PMID: 38269590 PMCID: PMC10880935 DOI: 10.1161/atvbaha.123.319516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND Single-cell RNA-Seq analysis can determine the heterogeneity of cells between different tissues at a single-cell level. Coronary artery endothelial cells (ECs) are important to coronary blood flow. However, little is known about the heterogeneity of coronary artery ECs, and cellular identity responses to flow. Identifying endothelial subpopulations will contribute to the precise localization of vascular endothelial subpopulations, thus enabling the precision of vascular injury treatment. METHODS Here, we performed a single-cell RNA sequencing of 31 962 cells and functional assays of 3 branches of the coronary arteries (right coronary artery/circumflex left coronary artery/anterior descending left coronary artery) in wild-type mice. RESULTS We found a compendium of 7 distinct cell types in mouse coronary arteries, mainly ECs, granulocytes, cardiac myocytes, smooth muscle cells, lymphocytes, myeloid cells, and fibroblast cells, and showed spatial heterogeneity between arterial branches. Furthermore, we revealed a subpopulation of coronary artery ECs, CD133+TRPV4high ECs. TRPV4 (transient receptor potential vanilloid 4) in CD133+TRPV4high ECs is important for regulating vasodilation and coronary blood flow. CONCLUSIONS Our study elucidates the nature and range of coronary arterial cell diversity and highlights the importance of coronary CD133+TRPV4high ECs in regulating coronary vascular tone.
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
- School of Food Science and Technology (A.M., D.H.), Jiangnan University, China
| | - Ka Zhang
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Hao Kan
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Mengru Gao
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Zhiwei Wang
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Tingting Zhou
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Jing Shao
- Wuxi School of Medicine (A.M., K.Z., H.K., M.G., Z.W., T.Z., J.S.), Jiangnan University, China
| | - Dongxu He
- School of Food Science and Technology (A.M., D.H.), Jiangnan University, China
| |
Collapse
|
10
|
Singh J, Jackson KL, Tang FS, Fu T, Nowell C, Salimova E, Kiriazis H, Ritchie RH, Head GA, Woodman OL, Qin CX. The pro-resolving mediator, annexin A1 regulates blood pressure, and age-associated changes in cardiovascular function and remodeling. FASEB J 2024; 38:e23457. [PMID: 38318648 DOI: 10.1096/fj.202301802r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/07/2024]
Abstract
Aging is associated with chronic, low-level inflammation which may contribute to cardiovascular pathologies such as hypertension and atherosclerosis. This chronic inflammation may be opposed by endogenous mechanisms to limit inflammation, for example, by the actions of annexin A1 (ANXA1), an endogenous glucocorticoid-regulated protein that has anti-inflammatory and pro-resolving activity. We hypothesized the pro-resolving mediator ANXA1 protects against age-induced changes in blood pressure (BP), cardiovascular structure and function, and cardiac senescence. BP was measured monthly in conscious mature (4-month) and middle-aged (12-month) ANXA1-deficient (ANXA1-/- ) and wild-type C57BL/6 mice. Body composition was measured using EchoMRI, and both cardiac and vascular function using ultrasound imaging. Cardiac hypertrophy, fibrosis and senescence, vascular fibrosis, elastin, and calcification were assessed histologically. Gene expression relevant to structural remodeling, inflammation, and cardiomyocyte senescence were also quantified. In C57BL/6 mice, progression from 4 to 12 months of age did not affect the majority of cardiovascular parameters measured, with the exception of mild cardiac hypertrophy, vascular calcium, and collagen deposition. Interestingly, ANXA1-/- mice exhibited higher BP, regardless of age. Additionally, age progression had a marked impact in ANXA1-/- mice, with markedly augmented vascular remodeling, impaired vascular distensibility, and body composition. Consistent with vascular dysfunction, cardiac dysfunction, and hypertrophy were also evident, together with markers of senescence and inflammation. These findings suggest that endogenous ANXA1 plays a critical role in regulating BP, cardiovascular function, and remodeling and delays cardiac senescence. Our findings support the development of novel ANXA1-based therapies to prevent age-related cardiovascular pathologies.
Collapse
Affiliation(s)
- Jaideep Singh
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Kristy L Jackson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Feng Shii Tang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ting Fu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ekaterina Salimova
- Monash Biomedical Imaging, Monash University, Clayton, Melbourne, Victoria, Australia
| | - Helen Kiriazis
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca H Ritchie
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Victoria, Australia
| | - Geoffrey A Head
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
| | - Owen L Woodman
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cheng Xue Qin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Baker Heart & Diabetes Institute, Melbourne, Victoria, Australia
- Department of Pharmacology, School of Pharmaceutical Sciences, Qilu College of Medicine, Shandong University, Jinan, China
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
11
|
Fan Q, Yan R, Li Y, Lu L, Liu J, Li S, Fu T, Xue Y, Liu J, Li Z. Exploring Immune Cell Diversity in the Lacrimal Glands of Healthy Mice: A Single-Cell RNA-Sequencing Atlas. Int J Mol Sci 2024; 25:1208. [PMID: 38279208 PMCID: PMC10816500 DOI: 10.3390/ijms25021208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/28/2024] Open
Abstract
The lacrimal gland is responsible for maintaining the health of the ocular surface through the production of tears. However, our understanding of the immune system within the lacrimal gland is currently limited. Therefore, in this study, we utilized single-cell RNA sequencing and bioinformatic analysis to identify and analyze immune cells and molecules present in the lacrimal glands of normal mice. A total of 34,891 cells were obtained from the lacrimal glands of mice and classified into 18 distinct cell clusters using Seurat clustering. Within these cell populations, 26 different immune cell subpopulations were identified, including T cells, innate lymphocytes, macrophages, mast cells, dendritic cells, and B cells. Network analysis revealed complex cell-cell interactions between these immune cells, with particularly significant interactions observed among T cells, macrophages, plasma cells, and dendritic cells. Interestingly, T cells were found to be the main source of ligands for the Thy1 signaling pathway, while M2 macrophages were identified as the primary target of this pathway. Moreover, some of these immune cells were validated using immunohistological techniques. Collectively, these findings highlight the abundance and interactions of immune cells and provide valuable insights into the complexity of the lacrimal gland immune system and its relevance to associated diseases.
Collapse
Affiliation(s)
- Qiwei Fan
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Q.F.); (J.L.)
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
| | - Ruyu Yan
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Yan Li
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Liyuan Lu
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Jiangman Liu
- Department of Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Q.F.); (J.L.)
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
| | - Senmao Li
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Ting Fu
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Yunxia Xue
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Jun Liu
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Zhijie Li
- International Ocular Surface Research Center, Key Laboratory for Regenerative Medicine, Institute of Ophthalmology, Jinan University, Guangzhou 510632, China; (R.Y.); (Y.L.); (L.L.); (S.L.); (T.F.); (Y.X.); (J.L.)
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| |
Collapse
|
12
|
Salazar-Martín AG, Kalluri AS, Villanueva MA, Hughes TK, Wadsworth MH, Dao TT, Balcells M, Nezami FR, Shalek AK, Edelman ER. Single-Cell RNA Sequencing Reveals That Adaptation of Human Aortic Endothelial Cells to Antiproliferative Therapies Is Modulated by Flow-Induced Shear Stress. Arterioscler Thromb Vasc Biol 2023; 43:2265-2281. [PMID: 37732484 PMCID: PMC10659257 DOI: 10.1161/atvbaha.123.319283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Endothelial cells (ECs) are capable of quickly responding in a coordinated manner to a wide array of stresses to maintain vascular homeostasis. Loss of EC cellular adaptation may be a potential marker for cardiovascular disease and a predictor of poor response to endovascular pharmacological interventions such as drug-eluting stents. Here, we report single-cell transcriptional profiling of ECs exposed to multiple stimulus classes to evaluate EC adaptation. METHODS Human aortic ECs were costimulated with both pathophysiological flows mimicking shear stress levels found in the human aorta (laminar and turbulent, ranging from 2.5 to 30 dynes/cm2) and clinically relevant antiproliferative drugs, namely paclitaxel and rapamycin. EC state in response to these stimuli was defined using single-cell RNA sequencing. RESULTS We identified differentially expressed genes and inferred the TF (transcription factor) landscape modulated by flow shear stress using single-cell RNA sequencing. These flow-sensitive markers differentiated previously identified spatially distinct subpopulations of ECs in the murine aorta. Moreover, distinct transcriptional modules defined flow- and drug-responsive EC adaptation singly and in combination. Flow shear stress was the dominant driver of EC state, altering their response to pharmacological therapies. CONCLUSIONS We showed that flow shear stress modulates the cellular capacity of ECs to respond to paclitaxel and rapamycin administration, suggesting that while responding to different flow patterns, ECs experience an impairment in their transcriptional adaptation to other stimuli.
Collapse
Affiliation(s)
- Antonio G. Salazar-Martín
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Broad Institute of MIT and Harvard, Cambridge, MA (A.G.S.-M., M.A.V., T.T.D., A.K.S.)
| | - Aditya S. Kalluri
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
| | - Martin A. Villanueva
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Broad Institute of MIT and Harvard, Cambridge, MA (A.G.S.-M., M.A.V., T.T.D., A.K.S.)
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA (M.A.V., T.K.H., M.H.W., T.T.D., A.K.S.)
- Departments of Biology (M.A.V.), Massachusetts Institute of Technology, Cambridge
| | - Travis K. Hughes
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Koch Institute for Integrative Cancer Research (T.K.H., M.H.W., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA (M.A.V., T.K.H., M.H.W., T.T.D., A.K.S.)
- Department of Immunology, Harvard Medical School, Boston, MA (T.K.H., M.H.W., A.K.S.)
| | - Marc H. Wadsworth
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Koch Institute for Integrative Cancer Research (T.K.H., M.H.W., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA (M.A.V., T.K.H., M.H.W., T.T.D., A.K.S.)
- Department of Immunology, Harvard Medical School, Boston, MA (T.K.H., M.H.W., A.K.S.)
| | - Tyler T. Dao
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Broad Institute of MIT and Harvard, Cambridge, MA (A.G.S.-M., M.A.V., T.T.D., A.K.S.)
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA (M.A.V., T.K.H., M.H.W., T.T.D., A.K.S.)
- Biological Engineering (T.T.D.), Massachusetts Institute of Technology, Cambridge
| | - Mercedes Balcells
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
| | - Farhad R. Nezami
- Division of Cardiac Surgery (F.R.N.), Brigham and Women’s Hospital, Boston, MA
| | - Alex K. Shalek
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Koch Institute for Integrative Cancer Research (T.K.H., M.H.W., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Broad Institute of MIT and Harvard, Cambridge, MA (A.G.S.-M., M.A.V., T.T.D., A.K.S.)
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA (M.A.V., T.K.H., M.H.W., T.T.D., A.K.S.)
- Chemistry (A.K.S.), Massachusetts Institute of Technology, Cambridge
- Department of Immunology, Harvard Medical School, Boston, MA (T.K.H., M.H.W., A.K.S.)
| | - Elazer R. Edelman
- Institute for Medical Engineering and Science (A.G.S.-M., A.S.K., M.A.V., T.K.H., M.H.W., T.T.D., M.B., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Koch Institute for Integrative Cancer Research (T.K.H., M.H.W., A.K.S., E.R.E.), Massachusetts Institute of Technology (MIT), Cambridge, MA
- Division of Cardiovascular Medicine, Department of Medicine (E.R.E.), Brigham and Women’s Hospital, Boston, MA
| |
Collapse
|
13
|
Arduini A, Fleming SJ, Xiao L, Hall AW, Akkad AD, Chaffin M, Bendinelli KJ, Tucker NR, Papangeli I, Mantineo H, Babadi M, Stegmann CM, García-Cardeña G, Lindsay ME, Klattenhoff C, Ellinor PT. Transcriptional profile of the rat cardiovascular system at single cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567085. [PMID: 38014050 PMCID: PMC10680727 DOI: 10.1101/2023.11.14.567085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Background Despite the critical role of the cardiovascular system, our understanding of its cellular and transcriptional diversity remains limited. We therefore sought to characterize the cellular composition, phenotypes, molecular pathways, and communication networks between cell types at the tissue and sub-tissue level across the cardiovascular system of the healthy Wistar rat, an important model in preclinical cardiovascular research. We obtained high quality tissue samples under controlled conditions that reveal a level of cellular detail so far inaccessible in human studies. Methods and Results We performed single nucleus RNA-sequencing in 78 samples in 10 distinct regions including the four chambers of the heart, ventricular septum, sinoatrial node, atrioventricular node, aorta, pulmonary artery, and pulmonary veins (PV), which produced an aggregate map of 505,835 nuclei. We identified 26 distinct cell types and additional subtypes, including a number of rare cell types such as PV cardiomyocytes and non-myelinating Schwann cells (NMSCs), and unique groups of vascular smooth muscle cells (VSMCs), endothelial cells (ECs) and fibroblasts (FBs), which gave rise to a detailed cell type distribution across tissues. We demonstrated differences in the cellular composition across different cardiac regions and tissue-specific differences in transcription for each cell type, highlighting the molecular diversity and complex tissue architecture of the cardiovascular system. Specifically, we observed great transcriptional heterogeneities among ECs and FBs. Importantly, several cell subtypes had a unique regional localization such as a subtype of VSMCs enriched in the large vasculature. We found the cellular makeup of PV tissue is closer to heart tissue than to the large arteries. We further explored the ligand-receptor repertoire across cell clusters and tissues, and observed tissue-enriched cellular communication networks, including heightened Nppa - Npr1/2/3 signaling in the sinoatrial node. Conclusions Through a large single nucleus sequencing effort encompassing over 500,000 nuclei, we broadened our understanding of cellular transcription in the healthy cardiovascular system. The existence of tissue-restricted cellular phenotypes suggests regional regulation of cardiovascular physiology. The overall conservation in gene expression and molecular pathways across rat and human cell types, together with our detailed transcriptional characterization of each cell type, offers the potential to identify novel therapeutic targets and improve preclinical models of cardiovascular disease.
Collapse
Affiliation(s)
- Alessandro Arduini
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
| | - Stephen J. Fleming
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA 02142
| | - Ling Xiao
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA 02114
| | - Amelia W. Hall
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA 02114
| | - Amer-Denis Akkad
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA, 02142
| | - Mark Chaffin
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
| | - Kayla J. Bendinelli
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
| | | | - Irinna Papangeli
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA, 02142
| | - Helene Mantineo
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA 02114
| | - Mehrtash Babadi
- Data Sciences Platform, The Broad Institute of MIT and Harvard, Cambridge, MA, USA 02142
| | | | - Guillermo García-Cardeña
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA 02215
| | - Mark E. Lindsay
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA 02114
| | | | - Patrick T. Ellinor
- Precision Cardiology Laboratory, The Broad Institute, Cambridge, MA, USA 02142
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA 02114
- Cardiology Division, Massachusetts General Hospital, Boston, MA, USA 02114
| |
Collapse
|
14
|
Abstract
Organismal aging exhibits wide-ranging hallmarks in divergent cell types across tissues, organs, and systems. The advancement of single-cell technologies and generation of rich datasets have afforded the scientific community the opportunity to decode these hallmarks of aging at an unprecedented scope and resolution. In this review, we describe the technological advancements and bioinformatic methodologies enabling data interpretation at the cellular level. Then, we outline the application of such technologies for decoding aging hallmarks and potential intervention targets and summarize common themes and context-specific molecular features in representative organ systems across the body. Finally, we provide a brief summary of available databases relevant for aging research and present an outlook on the opportunities in this emerging field.
Collapse
Affiliation(s)
- Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; ,
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Xu Chi
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China;
| | - Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; ,
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Zhejun Ji
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China;
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China;
- University of Chinese Academy of Sciences, Beijing, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; ,
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Advanced Innovation Center for Human Brain Protection and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China;
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Su X, Wang L, Ma N, Yang X, Liu C, Yang F, Li J, Yi X, Xing Y. Immune heterogeneity in cardiovascular diseases from a single-cell perspective. Front Cardiovasc Med 2023; 10:1057870. [PMID: 37180791 PMCID: PMC10167030 DOI: 10.3389/fcvm.2023.1057870] [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: 10/03/2022] [Accepted: 04/10/2023] [Indexed: 05/16/2023] Open
Abstract
A variety of immune cell subsets occupy different niches in the cardiovascular system, causing changes in the structure and function of the heart and vascular system, and driving the progress of cardiovascular diseases (CVDs). The immune cells infiltrating the injury site are highly diverse and integrate into a broad dynamic immune network that controls the dynamic changes of CVDs. Due to technical limitations, the effects and molecular mechanisms of these dynamic immune networks on CVDs have not been fully revealed. With recent advances in single-cell technologies such as single-cell RNA sequencing, systematic interrogation of the immune cell subsets is feasible and will provide insights into the way we understand the integrative behavior of immune populations. We no longer lightly ignore the role of individual cells, especially certain highly heterogeneous or rare subpopulations. We summarize the phenotypic diversity of immune cell subsets and their significance in three CVDs of atherosclerosis, myocardial ischemia and heart failure. We believe that such a review could enhance our understanding of how immune heterogeneity drives the progression of CVDs, help to elucidate the regulatory roles of immune cell subsets in disease, and thus guide the development of new immunotherapies.
Collapse
Affiliation(s)
- Xin Su
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Li Wang
- Department of Breast Surgery, Xingtai People’s Hospital, Xingtai, China
| | - Ning Ma
- Department of Breast Surgery, Dezhou Second People’s Hospital, Dezhou, China
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Can Liu
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Fan Yang
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Jun Li
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| | - Xin Yi
- Department of Cardiology, Beijing Huimin Hospital, Beijing, China
| | - Yanwei Xing
- China Academy of Chinese Medical Sciences, Guang’anmen Hospital, Beijing, China
| |
Collapse
|
16
|
DeRoo E, Zhou T, Yang H, Stranz A, Henke P, Liu B. A vein wall cell atlas of murine venous thrombosis determined by single-cell RNA sequencing. Commun Biol 2023; 6:130. [PMID: 36721040 PMCID: PMC9889765 DOI: 10.1038/s42003-023-04492-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
Deep vein thrombosis (DVT) is a common clinical problem, but its cellular and molecular mechanisms remain incompletely understood. In this study, we performed single-cell RNA sequencing on mouse inferior vena cava (IVC) 24 h after thrombus-inducing IVC ligation or sham operation. 9 cell types composed of multiple subpopulations were identified. Notable transcriptomic changes induced by DVT included a marked inflammatory response, elevated hypoxia, and globally reduced myogenesis. Analysis of individual cell populations revealed increased inflammation and reduced extracellular matrix production across smooth muscle cells and fibroblasts, juxtaposed against an early phenotypic shift in smooth muscle cell populations away from a contractile state. By characterizing the transcriptomic changes in the vein wall during acute venous thrombosis at the single-cell level, this work provides novel insights into early pathological events in the vein wall that may potentiate thrombus formation and result in long term adverse venous remodeling.
Collapse
Affiliation(s)
- Elise DeRoo
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Ting Zhou
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| | - Huan Yang
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Amelia Stranz
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Peter Henke
- Department of Surgery, Division of Vascular Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Bo Liu
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
- Department of Cellular and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, USA.
| |
Collapse
|
17
|
Alvino VV, Mohammed KAK, Gu Y, Madeddu P. Approaches for the isolation and long-term expansion of pericytes from human and animal tissues. Front Cardiovasc Med 2023; 9:1095141. [PMID: 36704463 PMCID: PMC9873410 DOI: 10.3389/fcvm.2022.1095141] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Pericytes surround capillaries in every organ of the human body. They are also present around the vasa vasorum, the small blood vessels that supply the walls of larger arteries and veins. The clinical interest in pericytes is rapidly growing, with the recognition of their crucial roles in controlling vascular function and possible therapeutic applications in regenerative medicine. Nonetheless, discrepancies in methods used to define, isolate, and expand pericytes are common and may affect reproducibility. Separating pure pericyte preparations from the continuum of perivascular mesenchymal cells is challenging. Moreover, variations in functional behavior and antigenic phenotype in response to environmental stimuli make it difficult to formulate an unequivocal definition of bona fide pericytes. Very few attempts were made to develop pericytes as a clinical-grade product. Therefore, this review is devoted to appraising current methodologies' pros and cons and proposing standardization and harmonization improvements. We highlight the importance of developing upgraded protocols to create therapeutic pericyte products according to the regulatory guidelines for clinical manufacturing. Finally, we describe how integrating RNA-seq techniques with single-cell spatial analysis, and functional assays may help realize the full potential of pericytes in health, disease, and tissue repair.
Collapse
Affiliation(s)
| | - Khaled Abdelsattar Kassem Mohammed
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Asyut, Egypt
| | - Yue Gu
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
18
|
Rodrigues Bento J, Meester J, Luyckx I, Peeters S, Verstraeten A, Loeys B. The Genetics and Typical Traits of Thoracic Aortic Aneurysm and Dissection. Annu Rev Genomics Hum Genet 2022; 23:223-253. [PMID: 36044906 DOI: 10.1146/annurev-genom-111521-104455] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genetic predisposition and risk factors such as hypertension and smoking can instigate the development of thoracic aortic aneurysm (TAA), which can lead to highly lethal aortic wall dissection and/or rupture. Monogenic defects in multiple genes involved in the elastin-contractile unit and the TGFβ signaling pathway have been associated with TAA in recent years, along with several genetic modifiers and risk-conferring polymorphisms. Advances in omics technology have also provided significant insights into the processes behind aortic wall degeneration: inflammation, epigenetics, vascular smooth muscle phenotype change and depletion, reactive oxygen species generation, mitochondrial dysfunction, and angiotensin signaling dysregulation. These recent advances and findings might pave the way for a therapy that is capable of stopping and perhaps even reversing aneurysm progression.
Collapse
Affiliation(s)
- Jotte Rodrigues Bento
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Josephina Meester
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Ilse Luyckx
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; .,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Silke Peeters
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Aline Verstraeten
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium;
| | - Bart Loeys
- Centre of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium; .,Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
19
|
Mussbacher M, Schossleitner K, Kral-Pointner JB, Salzmann M, Schrammel A, Schmid JA. More than Just a Monolayer: the Multifaceted Role of Endothelial Cells in the Pathophysiology of Atherosclerosis. Curr Atheroscler Rep 2022; 24:483-492. [PMID: 35404040 PMCID: PMC9162978 DOI: 10.1007/s11883-022-01023-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Purpose of the Review In this review, we summarize current insights into the versatile roles of endothelial cells in atherogenesis. Recent Findings The vascular endothelium represents the first barrier that prevents the entry of lipoproteins and leukocytes into the vessel wall, thereby controlling two key events in the pathogenesis of atherosclerosis. Disturbance of endothelial homeostasis increases vascular permeability, inflammation, and cellular trans-differentiation, which not only promotes the build-up of atherosclerotic plaques but is also involved in life-threatening thromboembolic complications such as plaque rupture and erosion. In this review, we focus on recent findings on endothelial lipoprotein transport, inflammation, cellular transitions, and barrier function. Summary By using cutting-edge technologies such as single-cell sequencing, epigenetics, and cell fate mapping, novel regulatory mechanisms and endothelial cell phenotypes have been discovered, which have not only challenged established concepts of endothelial activation, but have also led to a different view of the disease.
Collapse
Affiliation(s)
- Marion Mussbacher
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria.
| | - Klaudia Schossleitner
- Department of Dermatology, Skin and Endothelium Research Division, Medical University of Vienna, Vienna, Austria
| | - Julia B Kral-Pointner
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria.,Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria.,Department of Internal Medicine II/Cardiology, Medical University of Vienna, Vienna, Austria
| | - Astrid Schrammel
- Department of Pharmacology and Toxicology, University of Graz, Graz, Austria
| | - Johannes A Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University Vienna, Schwarzspanierstr. 17, 1090, Vienna, Austria.
| |
Collapse
|
20
|
Wu H, Hu T, Zhang L, Xia X, Liu X, Zhu Q, Wang M, Sun Z, Hao H, Cui Y, Parrish AR, Li DP, Hill MA, Xu C, Liu Z. Abdominal Aortic Endothelial Dysfunction Occurs in Female Mice With Dextran Sodium Sulfate-Induced Chronic Colitis Independently of Reactive Oxygen Species Formation. Front Cardiovasc Med 2022; 9:871335. [PMID: 35463755 PMCID: PMC9021429 DOI: 10.3389/fcvm.2022.871335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/04/2022] [Indexed: 01/19/2023] Open
Abstract
Background and Objective Inflammatory bowel disease (IBD) produces significant local and systemic inflammation with increased reactive oxygen species (ROS) formation. IBD Patients are at an increased risk for developing endothelial dysfunction and cardiovascular diseases. The present study tested the hypothesis that IBD impairs aortic endothelial function via ROS formation and investigate potential sex-related differences. Methods and Results Acute and chronic colitis models were induced in male and female C57BL/6 mice with dextran sodium sulfate (DSS) treatment. Aortic wall stiffness, endothelial function, and ROS levels, as well as serum levels of pro-inflammatory cytokines were evaluated. Acetylcholine (Ach)-induced endothelium-dependent relaxation of abdominal aorta without perivascular adipose tissue (PVAT) was significantly reduced in female mice, not males, with chronic colitis without a change in nitroglycerin-induced endothelium-independent relaxation. PVAT effectively preserved Ach-induced relaxation in abdominal aorta of female mice with chronic colitis. Aortic peak velocity, maximal intraluminal diameters, pulse wave velocity, distensibility and radial strain were preserved in mice with both acute and chronic colitis. Although pro-inflammatory cytokines levels were increased in mice with acute and chronic colitis, aortic ROS levels were not increased. Conclusion The data demonstrate that abdominal aortic endothelial function was attenuated selectively in female mice with chronic colitis independent of ROS formation. Further, PVAT played an important role in preserving endothelial function in female mice with chronic colitis.
Collapse
Affiliation(s)
- Hao Wu
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Tingzi Hu
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Linfang Zhang
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Xiujuan Xia
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Xuanyou Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Qiang Zhu
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Meifang Wang
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Yuqi Cui
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Alan R. Parrish
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO, United States
| | - De-Pei Li
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Canxia Xu
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| |
Collapse
|
21
|
Cheng CK, Lin X, Pu Y, Tse JKY, Wang Y, Zhang CL, Cao X, Lau CW, Huang J, He L, Luo JY, Shih YT, Wan S, Ng CF, Wang L, Ma RCW, Chiu JJ, Chan TF, Yu Tian X, Huang Y. SOX4 is a novel phenotypic regulator of endothelial cells in atherosclerosis revealed by single-cell analysis. J Adv Res 2022; 43:187-203. [PMID: 36585108 PMCID: PMC9811326 DOI: 10.1016/j.jare.2022.02.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 01/07/2023] Open
Abstract
INTRODUCTION Atherosclerotic complications represent the leading cause of cardiovascular mortality globally. Dysfunction of endothelial cells (ECs) often initiates the pathological events in atherosclerosis. OBJECTIVES In this study, we sought to investigate the transcriptional profile of atherosclerotic aortae, identify novel regulator in dysfunctional ECs and hence provide mechanistic insights into atherosclerotic progression. METHODS We applied single-cell RNA sequencing (scRNA-seq) on aortic cells from Western diet-fed apolipoprotein E-deficient (ApoE-/-) mice to explore the transcriptional landscape and heterogeneity of dysfunctional ECs. In vivo validation of SOX4 upregulation in ECs were performed in atherosclerotic tissues, including mouse aortic tissues, human coronary arteries, and human renal arteries. Single-cell analysis on human aortic aneurysmal tissue was also performed. Downstream vascular abnormalities induced by EC-specific SOX4 overexpression, and upstream modulators of SOX4 were revealed by biochemical assays, immunostaining, and wire myography. Effects of shear stress on endothelial SOX4 expression was investigated by in vitro hemodynamic study. RESULTS Among the compendium of aortic cells, mesenchymal markers in ECs were significantly enriched. Two EC subsets were subsequently distinguished, as the 'endothelial-like' and 'mesenchymal-like' subsets. Conventional assays consistently identified SOX4 as a novel atherosclerotic marker in mouse and different human arteries, additional to a cancer marker. EC-specific SOX4 overexpression promoted atherogenesis and endothelial-to-mesenchymal transition (EndoMT). Importantly, hyperlipidemia-associated cytokines and oscillatory blood flow upregulated, whereas the anti-diabetic drug metformin pharmacologically suppressed SOX4 level in ECs. CONCLUSION Our study unravels SOX4 as a novel phenotypic regulator during endothelial dysfunction, which exacerbates atherogenesis. Our study also pinpoints hyperlipidemia-associated cytokines and oscillatory blood flow as endogenous SOX4 inducers, providing more therapeutic insights against atherosclerotic diseases.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Department of Biomedical Sciences, City University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Xiao Lin
- School of Life Sciences, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Yujie Pu
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Joyce Ka Yu Tse
- School of Life Sciences, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Yu Wang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Cheng-Lin Zhang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Xiaoyun Cao
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Chi Wai Lau
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Juan Huang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Lei He
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Jiang-Yun Luo
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Song Wan
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Chi Fai Ng
- Department of Surgery, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Li Wang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Ronald Ching Wan Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan; School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Ting Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region
| | - Xiao Yu Tian
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region.
| | - Yu Huang
- School of Biomedical Sciences and Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Heart and Vascular Institute and Shenzhen Research Institute, The Chinese University of Hong Kong, 999077, Hong Kong Special Administrative Region; Department of Biomedical Sciences, City University of Hong Kong, 999077, Hong Kong Special Administrative Region.
| |
Collapse
|
22
|
Xie W, Ke Y, You Q, Li J, Chen L, Li D, Fang J, Chen X, Zhou Y, Chen L, Hong H. Single-Cell RNA Sequencing and Assay for Transposase-Accessible Chromatin Using Sequencing Reveals Cellular and Molecular Dynamics of Aortic Aging in Mice. Arterioscler Thromb Vasc Biol 2021; 42:156-171. [PMID: 34879708 DOI: 10.1161/atvbaha.121.316883] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE The impact of vascular aging on cardiovascular diseases has been extensively studied; however, little is known regarding the cellular and molecular mechanisms underlying age-related vascular aging in aortic cellular subpopulations. Approach and Results: Transcriptomes and transposase-accessible chromatin profiles from the aortas of 4-, 26-, and 86-week-old C57/BL6J mice were analyzed using single-cell RNA sequencing and assay for transposase-accessible chromatin sequencing. By integrating the heterogeneous transcriptome and chromatin accessibility data, we identified cell-specific TF (transcription factor) regulatory networks and open chromatin states. We also determined that aortic aging affects cell interactions, inflammation, cell type composition, dysregulation of transcriptional control, and chromatin accessibility. Endothelial cells 1 have higher gene set activity related to cellular senescence and aging than do endothelial cells 2. Moreover, construction of senescence trajectories shows that endothelial cell 1 and fibroblast senescence is associated with distinct TF open chromatin states and an mRNA expression model. CONCLUSIONS Our data provide a system-wide model for transcriptional and epigenetic regulation during aortic aging at single-cell resolution.
Collapse
Affiliation(s)
- Wenhui Xie
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yilang Ke
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qinyi You
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jing Li
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lu Chen
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Dang Li
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jun Fang
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaofeng Chen
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuanyuan Zhou
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Liangwan Chen
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huashan Hong
- Department of Geriatrics, Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fujian Institute of Geriatrics, Department of Cardiology, Department of Cardiac Surgery, Fujian Heart Disease Center, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
23
|
Mao A, Zhang P, Zhang K, Kan H, He D, Han X, Wang Z, Tang C, Ma X. Endothelial TRPV4-eNOS coupling as a vital therapy target for treatment of hypertension. Br J Pharmacol 2021; 179:2297-2312. [PMID: 34822720 DOI: 10.1111/bph.15755] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/25/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Reduced nitric oxide (NO) level and activity are signs of endothelial dysfunction, which is important in mediating blood pressure up-regulation. Previously, we demonstrated that transient receptor potential channel V4 (TRPV4) could form functional complex with other proteins to mediate vasodilation in the Endothelial cells (ECs). But how TRPV4 interacts with the NO pathway in larger arteries requires further exploration. EXPERIMENTAL APPROACH We used single-cell RNA-sequencing to find the CD106+ TRPV4high NOS3high ECs. The TRPV4-eNOS interaction was verified by co-immunoprecipitation and Immunofluorescence resonance energy transfer (FRET), and their binding site was found by site-directed mutagenesis. Endothelium-specific TRPV4 knockout (TRPV4EC -/- ) mice were used to study the effect of the TRPV4-eNOS interaction on blood pressure. A small molecule, JNc-463 was designed through molecular docking technology. KEY RESULTS We uncovered CD106+ TRPV4high NOS3high ECs in the mouse aorta, which they could regulate vasodilation via a TRPV4-eNOS interaction, and they were essential to regulate blood pressure. The TRPV4-eNOS interaction markedly decreased during the process of hypertension. We further attempted to identify the molecules re-join the TRPV4-eNOS interaction and develop a small-molecule drug, JNc-463, which could increase the TRPV4-eNOS interaction to enhance vasodilation, and exert antihypertensive effects in mice. CONCLUSION AND IMPLICATIONS This is the first study integrating single-cell RNA-Seq, single-cell functional study and drug screening in aorta. We identified a subpopulation of CD106+ TRPV4high NOS3high ECs, in which an impaired TRPV4-eNOS interaction was important in the progress of hypertension and we designed a small molecule, JNc-463 to improve the impaired TRPV4-eNOS interaction in hypertension.
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Peng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongxu He
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiping Han
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
24
|
Li Y, LeMaire SA, Shen YH. Molecular and Cellular Dynamics of Aortic Aneurysms Revealed by Single-Cell Transcriptomics. Arterioscler Thromb Vasc Biol 2021; 41:2671-2680. [PMID: 34615376 PMCID: PMC8556647 DOI: 10.1161/atvbaha.121.315852] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022]
Abstract
The aorta is highly heterogeneous, containing many different types of cells that perform sophisticated functions to maintain aortic homeostasis. Recently, single-cell RNA sequencing studies have provided substantial new insight into the heterogeneity of vascular cell types, the comprehensive molecular features of each cell type, and the phenotypic interrelationship between these cell populations. This new information has significantly improved our understanding of aortic biology and aneurysms at the molecular and cellular level. Here, we summarize these findings, with a focus on what single-cell RNA sequencing analysis has revealed about cellular heterogeneity, cellular transitions, communications among cell populations, and critical transcription factors in the vascular wall. We also review the information learned from single-cell RNA sequencing that has contributed to our understanding of the pathogenesis of vascular disease, such as the identification of cell types in which aneurysm-related genes and genetic variants function. Finally, we discuss the challenges and future directions of single-cell RNA sequencing applications in studies of aortic biology and diseases.
Collapse
Affiliation(s)
- Yanming Li
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | - Scott A LeMaire
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| | - Ying H Shen
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX
| |
Collapse
|
25
|
Kan H, Zhang K, Mao A, Geng L, Gao M, Feng L, You Q, Ma X. Single-cell transcriptome analysis reveals cellular heterogeneity in the ascending aortas of normal and high-fat diet-fed mice. Exp Mol Med 2021; 53:1379-1389. [PMID: 34548614 PMCID: PMC8492660 DOI: 10.1038/s12276-021-00671-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/13/2021] [Accepted: 07/21/2021] [Indexed: 01/26/2023] Open
Abstract
The aorta contains numerous cell types that contribute to vascular inflammation and thus the progression of aortic diseases. However, the heterogeneity and cellular composition of the ascending aorta in the setting of a high-fat diet (HFD) have not been fully assessed. We performed single-cell RNA sequencing on ascending aortas from mice fed a normal diet and mice fed a HFD. Unsupervised cluster analysis of the transcriptional profiles from 24,001 aortic cells identified 27 clusters representing 10 cell types: endothelial cells (ECs), fibroblasts, vascular smooth muscle cells (SMCs), immune cells (B cells, T cells, macrophages, and dendritic cells), mesothelial cells, pericytes, and neural cells. After HFD intake, subpopulations of endothelial cells with lipid transport and angiogenesis capacity and extensive expression of contractile genes were defined. In the HFD group, three major SMC subpopulations showed increased expression of extracellular matrix-degradation genes, and a synthetic SMC subcluster was proportionally increased. This increase was accompanied by upregulation of proinflammatory genes. Under HFD conditions, aortic-resident macrophage numbers were increased, and blood-derived macrophages showed the strongest expression of proinflammatory cytokines. Our study elucidates the nature and range of the cellular composition of the ascending aorta and increases understanding of the development and progression of aortic inflammatory disease.
Collapse
Affiliation(s)
- Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Li Geng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Mengru Gao
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Lei Feng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Qingjun You
- Department of Thoracic Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
26
|
Li F, Yan K, Wu L, Zheng Z, Du Y, Liu Z, Zhao L, Li W, Sheng Y, Ren L, Tang C, Zhu L. Single-cell RNA-seq reveals cellular heterogeneity of mouse carotid artery under disturbed flow. Cell Death Discov 2021; 7:180. [PMID: 34282126 PMCID: PMC8290019 DOI: 10.1038/s41420-021-00567-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/26/2021] [Accepted: 05/13/2021] [Indexed: 12/28/2022] Open
Abstract
Disturbed blood flow (d-flow) has been known to induce changes of the cells in the arterial wall, increasing the risk of atherosclerosis. However, the heterogeneity of the vascular cell populations under d-flow remains less understood. To generate d-flow in vivo, partial carotid artery ligation (PCL) was performed. Seven days after ligation, single-cell RNA sequencing of nine left carotid arteries (LCA) from the PCL group (10,262 cells) or control group (14,580 cells) was applied and a single-cell atlas of gene expression was constructed. The integrated analysis identified 15 distinct carotid cell clusters, including 10 d-flow-relevant subpopulations. Among endothelial cells, at least four subpopulations were identified, including Klk8hi ECs, Lrp1hi ECs, Dkk2hi ECs, and Cd36hi ECs. Analysis of GSVA and single-cell trajectories indicated that the previously undescribed Dkk2hi ECs subpopulation was mechanosensitive and potentially transformed from Klk8hi ECs under d-flow. D-flow-induced Spp1hi VSMCs subpopulation that appeared to be endowed with osteoblast differentiation, suggesting a role in arterial stiffness. Among the infiltrating cell subpopulations, Trem2hi Mφ, Birc5hi Mφ, DCs, CD4+ T cells, CXCR6+ T cells, NK cells, and granulocytes were identified under d-flow. Of note, the novel Birc5hi Mφ was identified as a potential contributor to the accumulation of macrophages in atherosclerosis. Finally, Dkk2hi ECs, and Cd36hi ECs were also found in the proatherosclerotic area of the aorta where the d-flow occurs. In conclusion, we presented a comprehensive single-cell atlas of all cells in the carotid artery under d-flow, identified previously unrecognized cell subpopulations and their gene expression signatures, and suggested their specialized functions.
Collapse
Affiliation(s)
- Fengchan Li
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Kunmin Yan
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Lili Wu
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Zhong Zheng
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Yun Du
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Ziting Liu
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Luyao Zhao
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Wei Li
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Yulan Sheng
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Lijie Ren
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China
| | - Chaojun Tang
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China.
- Collaborative Innovation Center of Hematology, Suzhou, Jiangsu, China.
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, Suzhou, Jiangsu, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Li Zhu
- Cyrus Tang Hematology Center, Suzhou, Jiangsu, China.
- Collaborative Innovation Center of Hematology, Suzhou, Jiangsu, China.
- Suzhou Key Laboratory of Thrombosis and Vascular Diseases, Suzhou, Jiangsu, China.
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
- State Key Laboratory of Radiation Medicine and Protection Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
27
|
Fu M, Song J. Single-Cell Transcriptomics Reveals the Cellular Heterogeneity of Cardiovascular Diseases. Front Cardiovasc Med 2021; 8:643519. [PMID: 34179129 PMCID: PMC8225933 DOI: 10.3389/fcvm.2021.643519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/13/2021] [Indexed: 12/23/2022] Open
Abstract
"A world in a wild flower, and a bodhi in a leaf," small cells contain huge secrets. The vasculature is composed of many multifunctional cell subpopulations, each of which is involved in the occurrence and development of cardiovascular diseases. Single-cell transcriptomics captures the full picture of genes expressed within individual cells, identifies rare or de novo cell subpopulations, analyzes single-cell trajectory and stem cell or progenitor cell lineage conversion, and compares healthy tissue and disease-related tissue at single-cell resolution. Single-cell transcriptomics has had a profound effect on the field of cardiovascular research over the past decade, as evidenced by the construction of cardiovascular cell landscape, as well as the clarification of cardiovascular diseases and the mechanism of stem cell or progenitor cell differentiation. The classification and proportion of cell subpopulations in vasculature vary with species, location, genotype, and disease, exhibiting unique gene expression characteristics in organ development, disease progression, and regression. Specific gene markers are expected to be the diagnostic criteria, therapeutic targets, or prognostic indicators of diseases. Therefore, treatment of vascular disease still has lots of potentials to develop. Herein, we summarize the cell clusters and gene expression patterns in normal vasculature and atherosclerosis, aortic aneurysm, and pulmonary hypertension to reveal vascular heterogeneity and new regulatory factors of cardiovascular disease in the use of single-cell transcriptomics and discuss its current limitations and promising clinical potential.
Collapse
Affiliation(s)
- Mengxia Fu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- The Cardiomyopathy Research Group at Fuwai Hospital, Beijing, China
- Department of Cardiovascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
28
|
Zhang CY, Hu YC, Zhang Y, Ma WD, Song YF, Quan XH, Guo X, Wang CX. Glutamine switches vascular smooth muscle cells to synthetic phenotype through inhibiting miR-143 expression and upregulating THY1 expression. Life Sci 2021; 277:119365. [PMID: 33741416 DOI: 10.1016/j.lfs.2021.119365] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/03/2021] [Accepted: 03/07/2021] [Indexed: 11/27/2022]
Abstract
AIMS Vascular smooth muscle cells (VSMCs) are involved in the pathogenesis of many human cardiovascular diseases. They modulate their phenotype from "contractile" to "synthetic" in response to changes in local environmental cues. How glutamine regulates the differentiation of VSMCs and the underlying mechanisms remain largely unknown. MAIN METHODS Here, we explored the effects of various doses of glutamine (0 mM, 1 mM, 2 mM, and 4 mM) on the proliferation, migration, and phenotypic switch of human VSMCs in vitro. Glutamine dose-dependently enhanced VSMC proliferation, and markedly increased VSMC migration. KEY FINDINGS Notably, glutamine promoted the phenotypic switch of VSMCs towards a synthetic phenotype, as evidenced by significantly decreased expression of contractile markers myosin heavy chain 11 (MYH11) and calponin while increased expression of synthetic markers collagen I and vimentin. Importantly, these changes upon glutamine treatments were attenuated after additional treatments with glutamine metabolism inhibitor BPTES. Additionally, glutamine downregulated miR-143 expression, and miR-143 inactivation alone resulted in enhanced proliferation, migration, and promoted the synthetic phenotype of VSMCs. Moreover, Thy-1 cell surface antigen (THY1) was validated as a downstream target of miR-143, and THY1 expression was upregulated by glutamine in VSMCs. Furthermore, either miR-143 overexpression or THY1 silencing abolished the effect of glutamine on proliferation, migration, and phenotypic switch of VSMCs, supporting a novel glutamine-miR-143-THY1 pathway in modulating VSMC functions. SIGNIFICANCE This study demonstrated a novel mechanism of glutamine in modulation of VSMC phenotypic switch by targeting miR-143 and THY1, and provides significant insight on targeted therapy of patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Chun-Yan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Yan-Chao Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Yan Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Wei-Dong Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Ya-Fan Song
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China
| | - Xiao-Hui Quan
- Department of Cardiovascular Medicine, Xi'an No.1 Hospital, 30 Fen Xiang, South Street, 710004 Xi'an, China
| | - Xuan Guo
- Department of Cardiovascular Medicine, Xi'an No.1 Hospital, 30 Fen Xiang, South Street, 710004 Xi'an, China
| | - Cong-Xia Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, 710004 Xi'an, China.
| |
Collapse
|
29
|
Wilson C, Zhang X, Lee MD, MacDonald M, Heathcote HR, Alorfi NMN, Buckley C, Dolan S, McCarron JG. Disrupted endothelial cell heterogeneity and network organization impair vascular function in prediabetic obesity. Metabolism 2020; 111:154340. [PMID: 32791171 PMCID: PMC7538703 DOI: 10.1016/j.metabol.2020.154340] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Obesity is a major risk factor for diabetes and cardiovascular diseases such as hypertension, heart failure, and stroke. Impaired endothelial function occurs in the earliest stages of obesity and underlies vascular alterations that give rise to cardiovascular disease. However, the mechanisms that link weight gain to endothelial dysfunction are ill-defined. Increasing evidence suggests that endothelial cells are not a population of uniform cells but are highly heterogeneous and are organized as a communicating multicellular network that controls vascular function. PURPOSE To investigate the hypothesis that disrupted endothelial heterogeneity and network-level organization contribute to impaired vascular reactivity in obesity. METHODS AND RESULTS To study obesity-related vascular function without complications associated with diabetes, a state of prediabetic obesity was induced in rats. Small artery diameter recordings confirmed nitric-oxide mediated vasodilator responses were dependent on increases in endothelial calcium levels and were impaired in obese animals. Single-photon imaging revealed a linear relationship between blood vessel relaxation and population-wide calcium responses. Obesity did not alter the slope of this relationship, but impaired calcium responses in the endothelial cell network. The network comprised structural and functional components. The structural architecture, a hexagonal lattice network of connected cells, was unchanged in obesity. The functional network contained sub-populations of clustered specialized agonist-sensing cells from which signals were communicated through the network. In obesity there were fewer but larger clusters of sensory cells and communication path lengths between clusters increased. Communication between neighboring cells was unaltered in obesity. Altered network organization resulted in impaired, population-level calcium signaling and deficient endothelial control of vascular tone. CONCLUSIONS The distribution of cells in the endothelial network is critical in determining overall vascular response. Altered cell heterogeneity and arrangement in obesity decreases endothelial function and provides a novel framework for understanding compromised endothelial function in cardiovascular disease.
Collapse
Affiliation(s)
- Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Margaret MacDonald
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Helen R Heathcote
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Nasser M N Alorfi
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Sharron Dolan
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|