1
|
Zhang X, Buckley C, Lee MD, Salaun C, MacDonald M, Wilson C, McCarron JG. Increased TRPV4 Channel Expression Enhances and Impairs Blood Vessel Function in Hypertension. Hypertension 2025; 82:57-68. [PMID: 39440451 DOI: 10.1161/hypertensionaha.124.23092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Endothelial cell TRPV4 (transient receptor potential vanilloid 4) channels provide a control point that is pivotal in regulating blood vessel diameter by mediating the Ca2+-dependent release of endothelial-derived vasoactive factors. In hypertension, TRPV4-mediated control of vascular function is disrupted, but the underlying mechanisms and precise physiological consequences remain controversial. METHODS Here, using a comprehensive array of methodologies, endothelial TRPV4 channel function was examined in intact mesenteric resistance arteries from normotensive Wistar-Kyoto and spontaneously hypertensive rats. RESULTS Our results show there is a notable shift in vascular reactivity in hypertension characterized by enhanced endothelium-dependent vasodilation at low levels of TRPV4 channel activation. However, at higher levels of TRPV4 activity, this vasodilatory response is reversed, contributing to the aberrant vascular tone observed in hypertension. The change in response, from dilation to constriction, was accompanied by a shift in intracellular Ca2+ signaling modalities arising from TRPV4 activity. Oscillatory TRPV4-evoked IP3 (inositol triphosphate)-mediated Ca2+ release, which underlies dilation, decreased, while the contraction inducing sustained Ca2+ rise, arising from TRPV4-mediated Ca2+ influx, increased. Our findings also reveal that while the sensitivity of endothelial cell TRPV4 to activation was unchanged, expression of the channel is upregulated and IP3 receptors are downregulated in hypertension. CONCLUSIONS These data highlight the intricate interplay between endothelial TRPV4 channel expression, intracellular Ca2+ signaling dynamics, and vascular reactivity. Moreover, the data support a new unifying hypothesis for the vascular impairment that accompanies hypertension. Specifically, endothelial cell TRPV4 channels play a dual role in modulating blood vessel function in hypertension.
Collapse
Affiliation(s)
- Xun Zhang
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Charlotte Buckley
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Matthew D Lee
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Christine Salaun
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Margaret MacDonald
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Calum Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
2
|
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
|
3
|
Zhang J, Liu S, Ding W, Wan J, Qin JJ, Wang M. Resolution of inflammation, an active process to restore the immune microenvironment balance: A novel drug target for treating arterial hypertension. Ageing Res Rev 2024; 99:102352. [PMID: 38857706 DOI: 10.1016/j.arr.2024.102352] [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: 11/24/2023] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024]
Abstract
The resolution of inflammation, the other side of the inflammatory response, is defined as an active and highly coordinated process that promotes the restoration of immune microenvironment balance and tissue repair. Inflammation resolution involves several key processes, including dampening proinflammatory signaling, specialized proresolving lipid mediator (SPM) production, nonlipid proresolving mediator production, efferocytosis and regulatory T-cell (Treg) induction. In recent years, increasing attention has been given to the effects of inflammation resolution on hypertension. Furthermore, our previous studies reported the antihypertensive effects of SPMs. Therefore, in this review, we aim to summarize and discuss the detailed association between arterial hypertension and inflammation resolution. Additional, the association between gut microbe-mediated immune and hypertension is discussed. This findings suggested that accelerating the resolution of inflammation can have beneficial effects on hypertension and its related organ damage. Exploring novel drug targets by focusing on various pathways involved in accelerating inflammation resolution will contribute to the treatment and control of hypertensive diseases in the future.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China; Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
4
|
Berillo O, Comeau K, Caillon A, Leclerc S, Shokoples BG, Mahmoud AUM, Andelfinger G, Paradis P, Schiffrin EL. CD28-expressing δ T cells are increased in perivascular adipose tissue of hypertensive mice and in subcutaneous adipose tissue of obese humans. J Hypertens 2024; 42:1256-1268. [PMID: 38704218 DOI: 10.1097/hjh.0000000000003725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2024]
Abstract
OBJECTIVES γδ T-lymphocytes play a role in angiotensin II (AngII)-induced hypertension, vascular injury and T-cell infiltration in perivascular adipose tissue (PVAT) in mice. Mesenteric arteries of hypertensive mice and subcutaneous arteries from obese humans present similar remodeling. We hypothesized that γδ T-cell subtypes in mesenteric vessels with PVAT (MV/PVAT) from hypertensive mice and subcutaneous adipose tissue (SAT) from obese humans, who are prone to develop hypertension, would be similar. METHODS Mice were infused with AngII for 14 days. MV/PVAT T-cells were used for single-cell RNA-sequencing (scRNA-seq). scRNA-seq data (GSE155960) of SAT CD45 + cells from three lean and three obese women were downloaded from the Gene Expression Omnibus database. RESULTS δ T-cell subclustering identified six δ T-cell subtypes. AngII increased T-cell receptor δ variable 4 ( Trdv4 ) + γδ T-effector memory cells and Cd28high δ T EM -cells, changes confirmed by flow cytometry. δ T-cell subclustering identified nine δ T-cell subtypes in human SAT. CD28 expressing δ T-cell subclustering demonstrated similar δ T-cell subpopulations in murine MV/PVAT and human SAT. Cd28+ γδ NKT EM and Cd28high δ T EM -cells increased in MV/PVAT from hypertensive mice and CD28high δ T EM -cells in SAT from obese women compared to the lean women. CONCLUSION Similar CD28 + δ T-cells were identified in murine MV/PVAT and human SAT. CD28 high δ T EM -cells increased in MV/PVAT in hypertensive mice and in SAT from humans with obesity, a prehypertensive condition. CD28 + δ T-lymphocytes could have a pathogenic role in human hypertension associated with obesity, and could be a potential target for therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Gregor Andelfinger
- Research Center, Sainte-Justine University Health Center
- Department of Pediatrics, University of Montreal, Montréal, Québec, Canada
| | | | - Ernesto L Schiffrin
- Lady Davis Institute for Medical Research
- Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University
| |
Collapse
|
5
|
Torimoto K, Elliott K, Nakayama Y, Yanagisawa H, Eguchi S. Cardiac and perivascular myofibroblasts, matrifibrocytes, and immune fibrocytes in hypertension; commonalities and differences with other cardiovascular diseases. Cardiovasc Res 2024; 120:567-580. [PMID: 38395029 PMCID: PMC11485269 DOI: 10.1093/cvr/cvae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/02/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertension is a major cause of cardiovascular diseases such as myocardial infarction and stroke. Cardiovascular fibrosis occurs with hypertension and contributes to vascular resistance, aortic stiffness, and cardiac hypertrophy. However, the molecular mechanisms leading to fibroblast activation in hypertension remain largely unknown. There are two types of fibrosis: replacement fibrosis and reactive fibrosis. Replacement fibrosis occurs in response to the loss of viable tissue to form a scar. Reactive fibrosis occurs in response to an increase in mechanical and neurohormonal stress. Although both types of fibrosis are considered adaptive processes, they become maladaptive when the tissue loss is too large, or the stress persists. Myofibroblasts represent a subpopulation of activated fibroblasts that have gained contractile function to promote wound healing. Therefore, myofibroblasts are a critical cell type that promotes replacement fibrosis. Although myofibroblasts were recognized as the fibroblasts participating in reactive fibrosis, recent experimental evidence indicated there are distinct fibroblast populations in cardiovascular reactive fibrosis. Accordingly, we will discuss the updated definition of fibroblast subpopulations, the regulatory mechanisms, and their potential roles in cardiovascular pathophysiology utilizing new knowledge from various lineage tracing and single-cell RNA sequencing studies. Among the fibroblast subpopulations, we will highlight the novel roles of matrifibrocytes and immune fibrocytes in cardiovascular fibrosis including experimental models of hypertension, pressure overload, myocardial infarction, atherosclerosis, aortic aneurysm, and nephrosclerosis. Exploration into the molecular mechanisms involved in the differentiation and activation of those fibroblast subpopulations may lead to novel treatments for end-organ damage associated with hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Keiichi Torimoto
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Katherine Elliott
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Yuki Nakayama
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Hiromi Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
- Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Satoru Eguchi
- Department of Cardiovascular Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| |
Collapse
|
6
|
Eguchi S, Torimoto K, Adebiyi A, Kanthakumar P, Bomfim GF, Wenceslau CF, Dahlen SA, Osei-Owusu P. Milestone Papers on Signal Transduction Mechanisms of Hypertension and Its Complications. Hypertension 2024; 81:977-990. [PMID: 38372140 PMCID: PMC11023792 DOI: 10.1161/hypertensionaha.123.21365] [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] [Indexed: 02/20/2024]
Abstract
To celebrate 100 years of American Heart Association-supported cardiovascular disease research, this review article highlights milestone papers that have significantly contributed to the current understanding of the signaling mechanisms driving hypertension and associated cardiovascular disorders. This article also includes a few of the future research directions arising from these critical findings. To accomplish this important mission, 4 principal investigators gathered their efforts to cover distinct yet intricately related areas of signaling mechanisms pertaining to the pathogenesis of hypertension. The renin-angiotensin system, canonical and novel contractile and vasodilatory pathways in the resistance vasculature, vascular smooth muscle regulation by membrane channels, and noncanonical regulation of blood pressure and vascular function will be described and discussed as major subjects.
Collapse
Affiliation(s)
- Satoru Eguchi
- Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University
| | - Keiichi Torimoto
- Department of Cardiovascular Science, Lewis Katz School of Medicine, Temple University
| | - Adebowale Adebiyi
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
| | - Praghalathan Kanthakumar
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
- Department of Anesthesiology and Perioperative Medicine, University of Missouri, Columbia, Missouri
- NextGen Precision Health, University of Missouri, Columbia, Missouri
| | - Gisele F. Bomfim
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Center, Department of Cell Biology and Anatomy, University of South Carolina School of Medicine
| | - Shelby A. Dahlen
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University
| | - Patrick Osei-Owusu
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University
| |
Collapse
|
7
|
Wu H, Zhai Y, Yu J, Wei L, Qi X. Transcriptome and proteome analyses reveal that upregulation of GSTM2 by allisartan improves cardiac remodeling and dysfunction in hypertensive rats. Exp Ther Med 2024; 27:220. [PMID: 38590561 PMCID: PMC11000455 DOI: 10.3892/etm.2024.12508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 02/20/2024] [Indexed: 04/10/2024] Open
Abstract
Long-term hypertension can lead to hypertensive heart disease, which ultimately progresses to heart failure. As an angiotensin receptor blocker antihypertensive drug, allisartan can control blood pressure, and improve cardiac remodeling and cardiac dysfunction caused by hypertension. The aim of the present study was to investigate the protective effects of allisartan on the heart of spontaneously hypertensive rats (SHRs) and the underlying mechanisms. SHRs were used as an animal model of hypertensive heart disease and were treated with allisartan orally at a dose of 25 mg/kg/day. The blood pressure levels of the rats were continuously monitored, their body and heart weights were measured, and their cardiac structure and function were evaluated using echocardiography. Wheat germ agglutinin staining and Masson trichrome staining were employed to assess the morphology of the myocardial tissue. In addition, transcriptome and proteome analyses were performed using the Solexa/Illumina sequencing platform and tandem mass tag technology, respectively. Immunofluorescence co-localization was conducted to analyze Nrf2 nuclear translocation, and TUNEL was performed to detect the levels of cell apoptosis. The protein expression levels of pro-collagen I, collagen III, phosphorylated (p)-AKT, AKT, p-PI3K and PI3K, and the mRNA expression levels of Col1a1 and Col3a1 were determined by western blotting and reverse transcription-quantitative PCR, respectively. Allisartan lowered blood pressure, attenuated cardiac remodeling and improved cardiac function in SHRs. In addition, allisartan alleviated cardiomyocyte hypertrophy and cardiac fibrosis. Allisartan also significantly affected the 'pentose phosphate pathway', 'fatty acid elongation', 'valine, leucine and isoleucine degradation', 'glutathione metabolism', and 'amino sugar and nucleotide sugar metabolism' pathways in the hearts of SHRs, and upregulated the expression levels of GSTM2. Furthermore, allisartan activated the PI3K-AKT-Nrf2 signaling pathway and inhibited cardiomyocyte apoptosis. In conclusion, the present study demonstrated that allisartan can effectively control blood pressure in SHRs, and improves cardiac remodeling and cardiac dysfunction. Allisartan may also upregulate the expression levels of GSTM2 in the hearts of SHRs and significantly affect glutathione metabolism, as determined by transcriptome and proteome analyses. The cardioprotective effect of allisartan may be mediated through activation of the PI3K-AKT-Nrf2 signaling pathway, upregulation of GSTM2 expression and reduction of cardiomyocyte apoptosis in SHRs.
Collapse
Affiliation(s)
- Hao Wu
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Yajun Zhai
- Graduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Jing Yu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Liping Wei
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| | - Xin Qi
- School of Medicine, Nankai University, Tianjin 300071, P.R. China
- Department of Cardiology, Tianjin Union Medical Center, Tianjin 300121, P.R. China
| |
Collapse
|
8
|
Adelus ML, Ding J, Tran BT, Conklin AC, Golebiewski AK, Stolze LK, Whalen MB, Cusanovich DA, Romanoski CE. Single-cell 'omic profiles of human aortic endothelial cells in vitro and human atherosclerotic lesions ex vivo reveal heterogeneity of endothelial subtype and response to activating perturbations. eLife 2024; 12:RP91729. [PMID: 38578680 PMCID: PMC10997331 DOI: 10.7554/elife.91729] [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] [Indexed: 04/06/2024] Open
Abstract
Heterogeneity in endothelial cell (EC) sub-phenotypes is becoming increasingly appreciated in atherosclerosis progression. Still, studies quantifying EC heterogeneity across whole transcriptomes and epigenomes in both in vitro and in vivo models are lacking. Multiomic profiling concurrently measuring transcriptomes and accessible chromatin in the same single cells was performed on six distinct primary cultures of human aortic ECs (HAECs) exposed to activating environments characteristic of the atherosclerotic microenvironment in vitro. Meta-analysis of single-cell transcriptomes across 17 human ex vivo arterial specimens was performed and two computational approaches quantitatively evaluated the similarity in molecular profiles between heterogeneous in vitro and ex vivo cell profiles. HAEC cultures were reproducibly populated by four major clusters with distinct pathway enrichment profiles and modest heterogeneous responses: EC1-angiogenic, EC2-proliferative, EC3-activated/mesenchymal-like, and EC4-mesenchymal. Quantitative comparisons between in vitro and ex vivo transcriptomes confirmed EC1 and EC2 as most canonically EC-like, and EC4 as most mesenchymal with minimal effects elicited by siERG and IL1B. Lastly, accessible chromatin regions unique to EC2 and EC4 were most enriched for coronary artery disease (CAD)-associated single-nucleotide polymorphisms from Genome Wide Association Studies (GWAS), suggesting that these cell phenotypes harbor CAD-modulating mechanisms. Primary EC cultures contain markedly heterogeneous cell subtypes defined by their molecular profiles. Surprisingly, the perturbations used here only modestly shifted cells between subpopulations, suggesting relatively stable molecular phenotypes in culture. Identifying consistently heterogeneous EC subpopulations between in vitro and ex vivo models should pave the way for improving in vitro systems while enabling the mechanisms governing heterogeneous cell state decisions.
Collapse
Affiliation(s)
- Maria L Adelus
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
- The Clinical Translational Sciences Graduate Program, The University of ArizonaTucsonUnited States
| | - Jiacheng Ding
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Binh T Tran
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Austin C Conklin
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Anna K Golebiewski
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Lindsey K Stolze
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Michael B Whalen
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Darren A Cusanovich
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
- Asthma and Airway Disease Research Center, The University of ArizonaTucsonUnited States
| | - Casey E Romanoski
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
- The Clinical Translational Sciences Graduate Program, The University of ArizonaTucsonUnited States
- Asthma and Airway Disease Research Center, The University of ArizonaTucsonUnited States
| |
Collapse
|
9
|
Sun X, Wu J, Zhang X, Xie C, Wei H, Li P, Yang Y, Yuan H, Cai J, Xiao Q, Cheng J, Xu Q. Atlas of Cell Repertoire Within Neointimal Lesions Is Metabolically Altered in Hypertensive Rats. Hypertension 2024; 81:787-800. [PMID: 38240164 DOI: 10.1161/hypertensionaha.123.22057] [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: 09/14/2023] [Accepted: 01/09/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND High blood pressure has been suggested to accelerate vascular injury-induced neointimal formation and progression. However, little is known about the intricate relationships between vascular injury and hypertension in the context of arterial remodeling. METHODS Single-cell RNA-sequencing analysis was used to depict the cell atlas of carotid arteries of Wistar Kyoto rats and spontaneously hypertensive rats with or without balloon injury. RESULTS We found that hypertension significantly aggravated balloon injury-induced arterial stenosis. A total of 36 202 cells from carotid arteries with or without balloon injury were included in single-cell RNA-sequencing analysis. Cell composition analysis showed that vascular injury and hypertension independently induced distinct aortic cell phenotypic alterations including immune cells, endothelial cells (ECs), and smooth muscle cells. Specifically, our data showed that injury and hypertension-induced specific EC phenotypic alterations, and revealed a transition from functional ECs to hypermetabolic, and eventually dysfunctional ECs in hypertensive rats upon balloon injury. Importantly, our data also showed that vascular injury and hypertension-induced different smooth muscle cell phenotypic alterations, characterized by deferential expression of synthetic signatures. Interestingly, pathway analysis showed that dysregulated metabolic pathways were a common feature in monocytes/macrophages, ECs, and smooth muscle cells in response to injury and hypertension. Functionally, we demonstrate that inhibition of mitochondrial respiration significantly ameliorated injury-induced neointimal formation in spontaneously hypertensive rats. CONCLUSIONS This study provides the cell landscape changes of the main aortic cell phenotypic alterations in response to injury and hypertension. Our findings suggest that targeting cellular mitochondrial respiration could be a novel therapeutic for patients with hypertension undergoing vascular angioplasty.
Collapse
Affiliation(s)
- Xiaolei Sun
- Department of General Surgery (Vascular Surgery), Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Luzhou, China (X.S., H.W.)
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Junru Wu
- Department of Cardiology and Center of Pharmacology, Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (J.W., H.Y., J. Cai)
| | - Xiaolin Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Cheng Xie
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Haijun Wei
- Department of General Surgery (Vascular Surgery), Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, The Affiliated Hospital of Southwest Medical University, Luzhou, China (X.S., H.W.)
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Hong Yuan
- Department of Cardiology and Center of Pharmacology, Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (J.W., H.Y., J. Cai)
| | - Jingjing Cai
- Department of Cardiology and Center of Pharmacology, Postdoctoral Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (J.W., H.Y., J. Cai)
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
| | - Qingbo Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Public Center of Experimental Technology, Southwest Medical University, Luzhou, China (X.S., X.Z., C.X., P.L., Y.Y., J. Cheng, Q. Xu)
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China (Q. Xu)
| |
Collapse
|
10
|
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
|
11
|
Liu X, Su M, Wei L, Zhang J, Wang W, Hao Q, Lin X, Wang L. Single-cell analysis of uterosacral ligament revealed cellular heterogeneity in women with pelvic organ prolapse. Commun Biol 2024; 7:159. [PMID: 38326542 PMCID: PMC10850063 DOI: 10.1038/s42003-024-05808-3] [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: 02/25/2023] [Accepted: 01/11/2024] [Indexed: 02/09/2024] Open
Abstract
Pelvic organ prolapse (POP) markedly affects the quality of life of women, including significant financial burden. Using single-cell RNA sequencing, we constructed a transcriptional profile of 30,452 single cells of the uterosacral ligament in POP and control samples, which has never been constructed before. We identified 10 major cell types, including smooth muscle cells, endothelial cells, fibroblasts, neutrophils, macrophages, monocytes, mast cells, T cells, B cells, and dendritic cells. We performed subpopulation analysis and pseudo-time analysis of POP primary cells, and explored differentially expressed genes. We verified previous cell clusters of human neutrophils of uterosacral ligaments. We found a significant reduction in receptor-ligand pairs related to ECM and cell adhesion between fibroblasts and endothelial cells in POP. The transcription factors related to the extracellular matrix, development, and immunity were identified in USL. Here we provide insight into the molecular mechanisms of POP and valuable information for future research directions.
Collapse
Affiliation(s)
- Xiaochun Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, China.
| | - Minna Su
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, China
| | - Lingyun Wei
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, China
| | - Jia Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, China
| | - Wenzhen Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, China
| | - Qian Hao
- Taiyuan Health School, 030012, Taiyuan, China
| | - Xiling Lin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 030032, Taiyuan, China
| | - Lili Wang
- Taiyuan University of Technology, 030024, Taiyuan, China
| |
Collapse
|
12
|
Khan SU, Huang Y, Ali H, Ali I, Ahmad S, Khan SU, Hussain T, Ullah M, Lu K. Single-cell RNA Sequencing (scRNA-seq): Advances and Challenges for Cardiovascular Diseases (CVDs). Curr Probl Cardiol 2024; 49:102202. [PMID: 37967800 DOI: 10.1016/j.cpcardiol.2023.102202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/11/2023] [Indexed: 11/17/2023]
Abstract
Implementing Single-cell RNA sequencing (scRNA-seq) has significantly enhanced our comprehension of cardiovascular diseases (CVDs), providing new opportunities to strengthen the prevention of CVDs progression. Cardiovascular diseases continue to be the primary cause of death worldwide. Improving treatment strategies and patient risk assessment requires a deeper understanding of the fundamental mechanisms underlying these disorders. The advanced and widespread use of Single-cell RNA sequencing enables a comprehensive investigation of the complex cellular makeup of the heart, surpassing essential descriptive aspects. This enhances our understanding of disease causes and directs functional research. The significant advancement in understanding cellular phenotypes has enhanced the study of fundamental cardiovascular science. scRNA-seq enables the identification of discrete cellular subgroups, unveiling previously unknown cell types in the heart and vascular systems that may have relevance to different disease pathologies. Moreover, scRNA-seq has revealed significant heterogeneity in phenotypes among distinct cell subtypes. Finally, we will examine current and upcoming scRNA-seq studies about various aspects of the cardiovascular system, assessing their potential impact on our understanding of the cardiovascular system and offering insight into how these technologies may revolutionise the diagnosis and treatment of cardiac conditions.
Collapse
Affiliation(s)
- Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China; Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China; Women Medical and Dental College, Khyber Medical University, Peshawar, KPK, 22020, Pakistan
| | - Yuqing Huang
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, China; Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Hamid Ali
- Department of Biosciences, COMSATS University Islamabad, Park Road Tarlai Kalan, Islamabad-44000
| | - Ijaz Ali
- Centre for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Hawally 32093, Kuwait
| | - Saleem Ahmad
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans 70112 LA, USA
| | - Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, People's Republic of China
| | - Talib Hussain
- Women Dental College Abbottabad, KPK, 22020, Pakistan
| | - Muneeb Ullah
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, KPK, Pakistan
| | - Kun Lu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing, 400715, China; Engineering Research Center of South Upland Agriculture, Ministry of Education, Chongqing, 400715, China.
| |
Collapse
|
13
|
Adelus ML, Ding J, Tran BT, Conklin AC, Golebiewski AK, Stolze LK, Whalen MB, Cusanovich DA, Romanoski CE. Single cell 'omic profiles of human aortic endothelial cells in vitro and human atherosclerotic lesions ex vivo reveals heterogeneity of endothelial subtype and response to activating perturbations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.03.535495. [PMID: 37066416 PMCID: PMC10104082 DOI: 10.1101/2023.04.03.535495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Objective Endothelial cells (ECs), macrophages, and vascular smooth muscle cells (VSMCs) are major cell types in atherosclerosis progression, and heterogeneity in EC sub-phenotypes are becoming increasingly appreciated. Still, studies quantifying EC heterogeneity across whole transcriptomes and epigenomes in both in vitro and in vivo models are lacking. Approach and Results To create an in vitro dataset to study human EC heterogeneity, multiomic profiling concurrently measuring transcriptomes and accessible chromatin in the same single cells was performed on six distinct primary cultures of human aortic ECs (HAECs). To model pro-inflammatory and activating environments characteristic of the atherosclerotic microenvironment in vitro, HAECs from at least three donors were exposed to three distinct perturbations with their respective controls: transforming growth factor beta-2 (TGFB2), interleukin-1 beta (IL1B), and siRNA-mediated knock-down of the endothelial transcription factor ERG (siERG). To form a comprehensive in vivo/ex vivo dataset of human atherosclerotic cell types, meta-analysis of single cell transcriptomes across 17 human arterial specimens was performed. Two computational approaches quantitatively evaluated the similarity in molecular profiles between heterogeneous in vitro and in vivo cell profiles. HAEC cultures were reproducibly populated by 4 major clusters with distinct pathway enrichment profiles: EC1-angiogenic, EC2-proliferative, EC3-activated/mesenchymal-like, and EC4-mesenchymal. Exposure to siERG, IL1B or TGFB2 elicited mostly distinct transcriptional and accessible chromatin responses. EC1 and EC2, the most canonically 'healthy' EC populations, were affected predominantly by siERG; the activated cluster EC3 was most responsive to IL1B; and the mesenchymal population EC4 was most affected by TGFB2. Quantitative comparisons between in vitro and in vivo transcriptomes confirmed EC1 and EC2 as most canonically EC-like, and EC4 as most mesenchymal with minimal effects elicited by siERG and IL1B. Lastly, accessible chromatin regions unique to EC2 and EC4 were most enriched for coronary artery disease (CAD)-associated SNPs from GWAS, suggesting these cell phenotypes harbor CAD-modulating mechanisms. Conclusion Primary EC cultures contain markedly heterogeneous cell subtypes defined by their molecular profiles. Surprisingly, the perturbations used here, which have been reported by others to be involved in the pathogenesis of atherosclerosis as well as induce endothelial-to-mesenchymal transition (EndMT), only modestly shifted cells between subpopulations, suggesting relatively stable molecular phenotypes in culture. Identifying consistently heterogeneous EC subpopulations between in vitro and in vivo models should pave the way for improving in vitro systems while enabling the mechanisms governing heterogeneous cell state decisions.
Collapse
Affiliation(s)
- Maria L. Adelus
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
- The Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jiacheng Ding
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Binh T. Tran
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Austin C. Conklin
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Anna K. Golebiewski
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Lindsey K. Stolze
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Michael B. Whalen
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Darren A. Cusanovich
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
- Asthma and Airway Disease Research Center, The University of Arizona, Tucson, AZ, 85721, USA
| | - Casey E. Romanoski
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
- The Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, 85721, USA
- Asthma and Airway Disease Research Center, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
14
|
Ren H, Dai R, Nik Nabil WN, Xi Z, Wang F, Xu H. Unveiling the dual role of autophagy in vascular remodelling and its related diseases. Biomed Pharmacother 2023; 168:115643. [PMID: 37839111 DOI: 10.1016/j.biopha.2023.115643] [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: 07/29/2023] [Revised: 09/18/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023] Open
Abstract
Vascular remodelling is an adaptive response to physiological and pathological stimuli that leads to structural and functional changes in the vascular intima, media, and adventitia. Pathological vascular remodelling is a hallmark feature of numerous vascular diseases, including atherosclerosis, hypertension, abdominal aortic aneurysm, pulmonary hypertension and preeclampsia. Autophagy is critical in maintaining cellular homeostasis, and its dysregulation has been implicated in the pathogenesis of various diseases, including vascular diseases. However, despite emerging evidence, the role of autophagy and its dual effects on vascular remodelling has garnered limited attention. Autophagy can exert protective and detrimental effects on the vascular intima, media and adventitia, thereby substantially influencing the course of vascular remodelling and its related vascular diseases. Currently, there has not been a review that thoroughly describes the regulation of autophagy in vascular remodelling and its impact on related diseases. Therefore, this review aimed to bridge this gap by focusing on the regulatory roles of autophagy in diseases related to vascular remodelling. This review also summarizes recent advancements in therapeutic agents targeting autophagy to regulate vascular remodelling. Additionally, this review offers an overview of recent breakthroughs in therapeutic agents targeting autophagy to regulate vascular remodelling. A deeper understanding of how autophagy orchestrates vascular remodelling can drive the development of targeted therapies for vascular diseases.
Collapse
Affiliation(s)
- Hangui Ren
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Wan Najbah Nik Nabil
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China; Pharmaceutical Services Program, Ministry of Health, Selangor 46200, Malaysia
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Feng Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China.
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China.
| |
Collapse
|
15
|
Sun X, Lu Y, Wu J, Wen Q, Li Z, Tang Y, Shi Y, He T, Liu L, Huang W, Weng C, Wu Q, Xiao Q, Yuan H, Xu Q, Cai J. Meta-Analysis of Single-Cell RNA-Seq Data Reveals the Mechanism of Formation and Heterogeneity of Tertiary Lymphoid Organ in Vascular Disease. Arterioscler Thromb Vasc Biol 2023; 43:1867-1886. [PMID: 37589134 PMCID: PMC10521807 DOI: 10.1161/atvbaha.123.318762] [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: 02/10/2023] [Accepted: 08/01/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND Tertiary lymphoid organs (TLOs) are ectopic lymphoid organs developed in nonlymphoid tissues with chronic inflammation, but little is known about their existence in different types of vascular diseases and the mechanism that mediated their development. METHODS To take advantage of single-cell RNA sequencing techniques, we integrated 28 single-cell RNA sequencing data sets containing 5 vascular disease models (atherosclerosis, abdominal aortic aneurysm, intimal hyperplasia, isograft, and allograft) to explore TLOs existence and environment supporting its growth systematically. We also searched Medline, Embase, PubMed, and Web of Science from inception to January 2022 for published histological images of vascular remodeling for histological evidence to support TLO genesis. RESULTS Accumulation and infiltration of innate and adaptive immune cells have been observed in various remodeling vessels. Interestingly, the proportion of such immune cells incrementally increases from atherosclerosis to intimal hyperplasia, abdominal aortic aneurysm, isograft, and allograft. Importantly, we uncovered that TLO structure cells, such as follicular helper T cells and germinal center B cells, present in all remodeled vessels. Among myeloid cells and lymphocytes, inflammatory macrophages, and T helper 17 cells are the major lymphoid tissue inducer cells which were found to be positively associated with the numbers of TLO structural cells in remodeled vessels. Vascular stromal cells also actively participate in vascular TLO genesis by communicating with myeloid cells and lymphocytes via CCLs (C-C motif chemokine ligands), CXCL (C-X-C motif ligand), lymphotoxin, BMP (bone morphogenetic protein) chemotactic, FGF-2 (fibroblast growth factor-2), and IGF (insulin growth factor) proliferation mechanisms, particularly for lymphoid tissue inducer cell aggregation. Additionally, the interaction between stromal cells and immune cells modulates extracellular matrix remodeling. Among TLO structure cells, follicular helper T, and germinal center B cells have strong interactions via TCR (T-cell receptor), CD40 (cluster of differentiation 40), and CXCL signaling, to promote the development and maturation of the germinal center in TLO. Consistently, by reviewing the histological images from the literature, TLO genesis was found in those vascular remodeling models. CONCLUSIONS Our analysis showed the existence of TLOs across 5 models of vascular diseases. The mechanisms that support TLOs formation in different models are heterogeneous. This study could be a valuable resource for understanding and discovering new therapeutic targets for various forms of vascular disease.
Collapse
Affiliation(s)
- Xuejing Sun
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Yao Lu
- The Center of Clinical Pharmacology (Y.L., H.Y.), Central South University, Changsha, China
| | - Junru Wu
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Qing Wen
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Zhengxin Li
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Yan Tang
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Yunmin Shi
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Tian He
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Lun Liu
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Wei Huang
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Chunyan Weng
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| | - Qing Wu
- The Third Xiangya Hospital and High-Performance Computing Center (Q. Wu), Central South University, Changsha, China
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
| | - Hong Yuan
- The Center of Clinical Pharmacology (Y.L., H.Y.), Central South University, Changsha, China
| | - Qingbo Xu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (Q. Xiao, Q. Xu)
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, China (Q. Xu)
| | - Jingjing Cai
- Department of Cardiology (X.S., J.W., Q. Wen, Z.L., Y.T., Y.S., T.H., L.L., W.H., C.W., J.C.), Central South University, Changsha, China
| |
Collapse
|
16
|
Hu Y, Cai Z, He B. Smooth Muscle Heterogeneity and Plasticity in Health and Aortic Aneurysmal Disease. Int J Mol Sci 2023; 24:11701. [PMID: 37511460 PMCID: PMC10380637 DOI: 10.3390/ijms241411701] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/16/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the medial layer of the aorta, which plays a critical role in the maintenance of aortic wall integrity. VSMCs have been suggested to have contractile and synthetic phenotypes and undergo phenotypic switching to contribute to the deteriorating aortic wall structure. Recently, the unprecedented heterogeneity and diversity of VSMCs and their complex relationship to aortic aneurysms (AAs) have been revealed by high-resolution research methods, such as lineage tracing and single-cell RNA sequencing. The aortic wall consists of VSMCs from different embryonic origins that respond unevenly to genetic defects that directly or indirectly regulate VSMC contractile phenotype. This difference predisposes to hereditary AAs in the aortic root and ascending aorta. Several VSMC phenotypes with different functions, for example, secreting VSMCs, proliferative VSMCs, mesenchymal stem cell-like VSMCs, immune-related VSMCs, proinflammatory VSMCs, senescent VSMCs, and stressed VSMCs are identified in non-hereditary AAs. The transformation of VSMCs into different phenotypes is an adaptive response to deleterious stimuli but can also trigger pathological remodeling that exacerbates the pathogenesis and development of AAs. This review is intended to contribute to the understanding of VSMC diversity in health and aneurysmal diseases. Papers that give an update on VSMC phenotype diversity in health and aneurysmal disease are summarized and recent insights on the role of VSMCs in AAs are discussed.
Collapse
Affiliation(s)
- Yunwen Hu
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| |
Collapse
|
17
|
Huyan Y, Wang C, Kang H, Chen X, Chang Y, Liu S, Song J. Single-Cell Transcriptome Sequencing Reveals Molecular Mechanisms of Renal Injury in Essential Hypertension. Kidney Blood Press Res 2023; 48:297-313. [PMID: 37062270 PMCID: PMC10308540 DOI: 10.1159/000530624] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
INTRODUCTION Hypertensive nephropathy is characterized by glomerular and tubulointerstitial damage, but we know little about changes in cell-specific gene expression in the early stages of hypertensive kidney injury, which usually has no obvious pathological changes. METHODS We performed unbiased single-cell RNA sequencing of rat kidney samples from hypertensive kidney injury to generate 10,602 single-cell transcriptomes from 2 control and 2 early stage hypertensive kidney injury samples. RESULTS All major cell types of the kidney were represented in the final dataset. Side-by-side comparisons showed that cell type-specific changes in gene expression are critical for functional impairment of glomeruli and tubules and activation of immune cells. In particular, we found a significantly reduced gene expression profile of maintaining vascular integrity in glomerular cells of hypertensive kidney injury. Meanwhile, the expression of genes associated with oxidative stress injury and fibrosis in the renal tubules and collecting ducts was elevated, but the degree of tubular cells response to injury differed between parts. We also found a signature of immune cell infiltration in hypertensive kidney injury. CONCLUSION Exploring the changes of gene expression in hypertension-injured kidneys may be helpful to identify the early biomarkers and signal pathways of this disease. Our data provide rich resources for understanding the pathogenesis of hypertensive renal injury and formulating effective treatment strategies.
Collapse
Affiliation(s)
- Yige Huyan
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunyue Wang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongen Kang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Xiao Chen
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Chang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sheng Liu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangping Song
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Long X, Yuan X, Du J. Single-cell and spatial transcriptomics: Advances in heart development and disease applications. Comput Struct Biotechnol J 2023; 21:2717-2731. [PMID: 37181659 PMCID: PMC10173363 DOI: 10.1016/j.csbj.2023.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Current transcriptomics technologies, including bulk RNA-seq, single-cell RNA sequencing (scRNA-seq), single-nucleus RNA-sequencing (snRNA-seq), and spatial transcriptomics (ST), provide novel insights into the spatial and temporal dynamics of gene expression during cardiac development and disease processes. Cardiac development is a highly sophisticated process involving the regulation of numerous key genes and signaling pathways at specific anatomical sites and developmental stages. Exploring the cell biological mechanisms involved in cardiogenesis also contributes to congenital heart disease research. Meanwhile, the severity of distinct heart diseases, such as coronary heart disease, valvular disease, cardiomyopathy, and heart failure, is associated with cellular transcriptional heterogeneity and phenotypic alteration. Integrating transcriptomic technologies in the clinical diagnosis and treatment of heart diseases will aid in advancing precision medicine. In this review, we summarize applications of scRNA-seq and ST in the cardiac field, including organogenesis and clinical diseases, and provide insights into the promise of single-cell and spatial transcriptomics in translational research and precision medicine.
Collapse
Affiliation(s)
- Xianglin Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
19
|
Anderson JR, Morin EE, Brayer KJ, Salbato S, Gonzalez Bosc LV, Kanagy NL, Naik JS. Single-cell transcriptomic heterogeneity between conduit and resistance mesenteric arteries in rats. Physiol Genomics 2023; 55:179-193. [PMID: 36912534 PMCID: PMC10085562 DOI: 10.1152/physiolgenomics.00126.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
The endothelium contains morphologically similar cells throughout the vasculature, but individual cells along the length of a single vascular tree or in different regional circulations function dissimilarly. When observations made in large arteries are extrapolated to explain the function of endothelial cells (ECs) in the resistance vasculature, only a fraction of these observations are consistent between artery sizes. To what extent endothelial (EC) and vascular smooth muscle cells (VSMCs) from different arteriolar segments of the same tissue differ phenotypically at the single-cell level remains unknown. Therefore, single-cell RNA-seq (10x Genomics) was performed using a 10X Genomics Chromium system. Cells were enzymatically digested from large (>300 µm) and small (<150 µm) mesenteric arteries from nine adult male Sprague-Dawley rats, pooled to create six samples (3 rats/sample, 3 samples/group). After normalized integration, the dataset was scaled before unsupervised cell clustering and cluster visualization using UMAP plots. Differential gene expression analysis allowed us to infer the biological identity of different clusters. Our analysis revealed 630 and 641 differentially expressed genes (DEGs) between conduit and resistance arteries for ECs and VSMCs, respectively. Gene ontology analysis (GO-Biological Processes, GOBP) of scRNA-seq data discovered 562 and 270 pathways for ECs and VSMCs, respectively, that differed between large and small arteries. We identified eight and seven unique ECs and VSMCs subpopulations, respectively, with DEGs and pathways identified for each cluster. These results and this dataset allow the discovery and support of novel hypotheses needed to identify mechanisms that determine the phenotypic heterogeneity between conduit and resistance arteries.
Collapse
Affiliation(s)
- Jacob R Anderson
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Emily E Morin
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Kathryn J Brayer
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Sophia Salbato
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Laura V Gonzalez Bosc
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Nancy L Kanagy
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| | - Jay S Naik
- Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States
| |
Collapse
|
20
|
Díaz Del Campo LS, García-Redondo AB, Rodríguez C, Zaragoza C, Duro-Sánchez S, Palmas F, de Benito-Bueno A, Socuéllamos PG, Peraza DA, Rodrigues-Díez R, Valenzuela C, Dalli J, Salaices M, Briones AM. Resolvin D2 Attenuates Cardiovascular Damage in Angiotensin II-Induced Hypertension. Hypertension 2023; 80:84-96. [PMID: 36337053 DOI: 10.1161/hypertensionaha.122.19448] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Resolution of inflammation is orchestrated by specialized proresolving lipid mediators (SPMs), and this would be impaired in some cardiovascular diseases. Among SPMs, resolvins (Rv) have beneficial effects in cardiovascular pathologies, but little is known about their effect on cardiovascular damage in hypertension. METHODS Aorta, small mesenteric arteries, heart, and peritoneal macrophages were taken from C57BL/6J mice, infused or not with angiotensin II (AngII; 1.44 mg/kg/day, 14 days) in presence or absence of resolvin D2 (RvD2) (100 ng/mice, every second day) starting 1 day before or 7 days after AngII infusion. RESULTS Enzymes and receptors involved in SPMs biosynthesis and signaling were increased in aorta or heart from AngII-infused mice. We also observed a differential regulation of SPMs in heart from these mice. Preventive treatment with RvD2 partially avoided AngII-induced hypertension and protected the heart and large and small vessels against functional and structural alterations induced by AngII. RvD2 increased the availability of vasoprotective factors, modified SPMs profile, decreased cardiovascular fibrosis, and increased the infiltration of pro-resolving macrophages. When administered in hypertensive animals with established cardiovascular damage, RvD2 partially improved cardiovascular function and structure, decreased fibrosis, reduced the infiltration of neutrophils, and shifted macrophage phenotype toward a pro-resolving phenotype. CONCLUSIONS There is a disbalance between proinflammatory and resolution mediators in hypertension. RvD2 protects cardiovascular function and structure when administered before and after the development of hypertension by modulating vascular factors, fibrosis and inflammation. Activating resolution mechanisms by treatment with RvD2 may represent a novel therapeutic strategy for the treatment of hypertensive cardiovascular disease.
Collapse
Affiliation(s)
- Lucia S Díaz Del Campo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (L.S.D.d.C., A.B.G.-R., S.D.-S, R.R.-D., M.S., A.M.B.)
| | - Ana B García-Redondo
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (L.S.D.d.C., A.B.G.-R., S.D.-S, R.R.-D., M.S., A.M.B.).,Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (A.B.G.-R.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.B.G.-R., R.R.-D, M.S., A.M.B.).,CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.)
| | - Cristina Rodríguez
- CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.).,Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain (C.R.)
| | - Carlos Zaragoza
- CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.).,Unidad de Investigación Cardiovascular, Departamento de Cardiología, Hospital Ramón y Cajal (IRYCIS), Universidad Francisco de Vitoria, Madrid, Spain (C.Z.)
| | - Santiago Duro-Sánchez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (L.S.D.d.C., A.B.G.-R., S.D.-S, R.R.-D., M.S., A.M.B.)
| | - Francesco Palmas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom (F.P., J.D.)
| | - Angela de Benito-Bueno
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom (J.D.)
| | - Paula G Socuéllamos
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain (A.d.B.-B., P.G.S., D.A.P., C.V.)
| | - Diego A Peraza
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain (A.d.B.-B., P.G.S., D.A.P., C.V.)
| | - Raquel Rodrigues-Díez
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (L.S.D.d.C., A.B.G.-R., S.D.-S, R.R.-D., M.S., A.M.B.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.B.G.-R., R.R.-D, M.S., A.M.B.).,CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.)
| | - Carmen Valenzuela
- CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.).,Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain (A.d.B.-B., P.G.S., D.A.P., C.V.)
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom (F.P., J.D.).,Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom (J.D.)
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (L.S.D.d.C., A.B.G.-R., S.D.-S, R.R.-D., M.S., A.M.B.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.B.G.-R., R.R.-D, M.S., A.M.B.).,CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.)
| | - Ana M Briones
- Departamento de Farmacología, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain (L.S.D.d.C., A.B.G.-R., S.D.-S, R.R.-D., M.S., A.M.B.).,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.B.G.-R., R.R.-D, M.S., A.M.B.).,CIBER Cardiovascular, Spain (A.B.G.-R., C.R., C.Z., R.R.-D., C.V., M.S., A.M.B.)
| |
Collapse
|
21
|
Fang Q, Wang J, Wei J, Long X, Wang Y, He J, Yuan X, Du J. Transcriptomic profile analysis of the left atrium in spontaneously hypertensive rats in the early stage. Front Pharmacol 2022; 13:989636. [PMID: 36324689 PMCID: PMC9620422 DOI: 10.3389/fphar.2022.989636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/28/2022] [Indexed: 11/19/2022] Open
Abstract
Left atrial remodeling, characterized by enlargement and hypertrophy of the left atrium and increased fibrosis, was accompanied by an increased incidence of atrial fibrillation. While before morphological changes at the early stage of hypertension, how overloaded hypertension influences the transcriptomic profile of the left atrium remains unclear. Therefore, RNA-sequencing was performed to define the RNA expressing profiles of left atrium in spontaneously hypertensive rats (SHRs) and normotensive Wistar-Kyoto (WKY) rats as a control group. We also compared the changes in the RNA expression profiles in SHRs treated with an angiotensin receptor blocker (ARB) and angiotensin receptor-neprilysin inhibitor (ARNI) to assess the distinct effects on the left atrium. In total, 1,558 differentially expressed genes were found in the left atrium between WKY rats and SHRs. Bioinformatics analysis showed that these mRNAs could regulate upstream pathways in atrial remodeling through atrial fibrosis, inflammation, electrical remodeling, and cardiac metabolism. The regulated transcripts detected in the left atrial tissue in both the ARB-treated and ARNI-treated groups were related to metabolism. In contrast to the ARB-treated rates, the transcripts in ARNI-treated rats were mapped to the cyclic guanosine monophosphate-protein kinase G signaling pathway.
Collapse
Affiliation(s)
- Qinghua Fang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiangjun Wei
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xianglin Long
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiacheng He
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Yuan
- Department of Nephrology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jianlin Du
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Jianlin Du,
| |
Collapse
|
22
|
Ke Y, Jian-yuan H, Ping Z, Yue W, Na X, Jian Y, Kai-xuan L, Yi-fan S, Han-bin L, Rong L. The progressive application of single-cell RNA sequencing technology in cardiovascular diseases. Biomed Pharmacother 2022; 154:113604. [DOI: 10.1016/j.biopha.2022.113604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/20/2022] [Accepted: 08/23/2022] [Indexed: 11/02/2022] Open
|
23
|
Wahiduzzaman M, Liu Y, Huang T, Wei W, Li Y. Cell-cell communication analysis for single-cell RNA sequencing and its applications in carcinogenesis and COVID-19. BIOSAFETY AND HEALTH 2022. [DOI: 10.1016/j.bsheal.2022.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
24
|
The Neuronal Transcription Factor Creb3l1 Potential Upregulates Ntrk2 in the Hypertensive Microenvironment to Promote Vascular Smooth Muscle Cell-Neuron Interaction and Prevent Neurons from Ferroptosis: A Bioinformatic Research of scRNA-seq Data. DISEASE MARKERS 2022; 2022:8339759. [PMID: 35186168 PMCID: PMC8853773 DOI: 10.1155/2022/8339759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022]
Abstract
Background There is still a lack of knowledge regarding the association between hypertension and ferroptosis. A single-cell approach was used to study the changes in neuropeptide expression as they might contribute to the mechanisms leading to ferroptosis in a hypertensive microenvironment. Methods We analyzed 11798 cells from the SHR group and 12589 cells from the WKY group of mouse arterial cells. CellPhoneDB was used for cell communication analysis, and the SCENIC method was used to identify key transcription factors in neurons. The correlation between Ntrk2 and ferroptosis-related genes was further analyzed and validated via quantitative polymerase chain reaction. Results The arterial cells were clustered into six cell types. Ligand-receptor analysis suggested that Ngf, Ntf3, Cxcr4, and Ntrk2 were key neuropeptide-related genes involved in the communication between vascular smooth muscle cells and neural cells. In the hypertensive microenvironment, the neuronal transcription factor Creb3l1 appears to play a key role in the upregulation of Ntrk2 to promote the interaction between neurons and vascular smooth muscle cells. An association between Ntrk2 and the ferroptosis death inhibitor Gpx4 was suggested. RT-qPCR experiments confirmed that Ntrk2 downregulation in neural cells was followed by downregulated expression of Gpx4. Conclusions Creb3l1, a key transcription factor in vascular neurons, may upregulate Ntrk2 to promote vascular smooth muscle cell-neuron interaction and thereby potentially prevent ferroptosis in neurons.
Collapse
|
25
|
Zhang M, Che C, Cheng J, Li P, Yang Y. Ion channels in stem cells and their roles in stem cell biology and vascular diseases. J Mol Cell Cardiol 2022; 166:63-73. [PMID: 35143836 DOI: 10.1016/j.yjmcc.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
Abstract
Stem cell therapy may be a promising option for the treatment of vascular diseases. In recent years, significant progress has been made in stem cell research, especially in the mechanism of stem cell activation, homing and differentiation in vascular repair and reconstruction. Current research on stem cells focuses on protein expression and transcriptional networks. Ion channels are considered to be the basis for the generation of bioelectrical signals, which control the proliferation, differentiation and migration of various cell types. Although heterogeneity of multiple ion channels has been found in different types of stem cells, it is unclear whether the heterogeneous expression of ion channels is related to different cell subpopulations and/or different stages of the cell cycle. There is still a long way to go in clinical treatment by using the regulation of stem cell ion channels. In this review, we reviewed the main ion channels found on stem cells, their expression and function in stem cell proliferation, differentiation and migration, and the research status of stem cells' involvement in vascular diseases.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Chang Che
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| |
Collapse
|
26
|
Gupta RM, Plutzky J. The Aging Aorta: Are We Only as Old as Our Endothelium? Arterioscler Thromb Vasc Biol 2022; 42:172-174. [PMID: 35080899 PMCID: PMC9741947 DOI: 10.1161/atvbaha.121.317275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Rajat M. Gupta
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA, USA,Division of Genetics, Brigham and Women’s Hospital, Boston, MA, USA,Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jorge Plutzky
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
27
|
Rizzoni D, De Ciuceis C, Szczepaniak P, Paradis P, Schiffrin EL, Guzik TJ. Immune System and Microvascular Remodeling in Humans. Hypertension 2022; 79:691-705. [PMID: 35098718 DOI: 10.1161/hypertensionaha.121.17955] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Low-grade inflammatory processes and related oxidative stress may have a key role in the pathogenesis of hypertension and hypertension-mediated organ damage. Innate immune cells, such as neutrophils, dendritic cells, monocytes/macrophages, as well as unconventional T lymphocytes like γδ T cells contribute to hypertension and may trigger vascular inflammation. Adaptive immunity has been demonstrated to participate in elevation of blood pressure and in vascular and kidney injury. In particular, effector T lymphocytes (Th1, Th2, and Th17) may play a relevant role in promoting hypertension and microvascular remodeling, whereas T-regulatory lymphocytes may have a protective role. Effector cytokines produced by these immune cells lead to increased oxidative stress, endothelial dysfunction and contribute to target organ damage in hypertension. A possible role of immune cell subpopulations in the development and regression of microvascular remodeling has also been proposed in humans with hypertension. The present review summarizes the key immune mechanisms that may participate in the pathophysiology of hypertension-mediated inflammation and vascular remodeling; advances in this field may provide the basis for novel therapeutics for hypertension.
Collapse
Affiliation(s)
- Damiano Rizzoni
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Italy (D.R., C.D.C.).,Division of Medicine, Spedali Civili di Brescia, Montichiari, Italy (D.R.)
| | - Carolina De Ciuceis
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Italy (D.R., C.D.C.)
| | - Piotr Szczepaniak
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (P.S., T.J.G.).,Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (P.S., T.J.G.)
| | - Pierre Paradis
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Montreal, Québec, Canada (P.P., E.L.S.)
| | - Ernesto L Schiffrin
- Hypertension and Vascular Research Unit, Lady Davis Institute for Medical Research, Montreal, Québec, Canada (P.P., E.L.S.).,Department of Medicine, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, Québec, Canada (E.L.S.)
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom (P.S., T.J.G.).,Department of Medicine, Jagiellonian University Medical College, Krakow, Poland (P.S., T.J.G.)
| |
Collapse
|
28
|
Fan Y, Zhou H, Liu X, Li J, Xu K, Fu X, Ye L, Li G. Applications of Single-Cell RNA Sequencing in Cardiovascular Research. Front Cell Dev Biol 2022; 9:810232. [PMID: 35174168 PMCID: PMC8841340 DOI: 10.3389/fcell.2021.810232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022] Open
Abstract
In recent years, cardiovascular disease (CVD) continues to be the leading cause of global disease burden. Extensive efforts have been made across basic, translational, and clinical research domains to curb the CVD epidemic and improve the health of the population. The successful completion of the Human Genome Project catapulted sequencing technology into the mainstream and aroused the interests of clinicians and scientific researchers alike. Advances in single-cell RNA sequencing (scRNA-seq), which is based on the transcriptional phenotypes of individual cells, have enabled the investigation of cellular fate, heterogeneity, and cell–cell interactions, as well as cell lineage determination, at a single-cell resolution. In this review, we summarize recent findings on the embryological development of the cardiovascular system and the pathogenesis and treatment of cardiovascular disease, as revealed by scRNA-seq technology. In particular, we discuss how scRNA-seq can help identify potential targets for the treatment of cardiovascular diseases and conclude with future perspectives for scRNA-seq.
Collapse
Affiliation(s)
- Yu Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- Department of Obstetrics, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Han Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xuexue Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jingyan Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ke Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiaodong Fu
- Department of Obstetrics, Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Ye
- National Heart Research Institute of Singapore, Singapore, Singapore
- *Correspondence: Lei Ye, ; Guang Li,
| | - Guang Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
- *Correspondence: Lei Ye, ; Guang Li,
| |
Collapse
|
29
|
Catar R, Herse-Naether M, Zhu N, Wagner P, Wischnewski O, Kusch A, Kamhieh-Milz J, Eisenreich A, Rauch U, Hegner B, Heidecke H, Kill A, Riemekasten G, Kleinau G, Scheerer P, Dragun D, Philippe A. Autoantibodies Targeting AT 1- and ET A-Receptors Link Endothelial Proliferation and Coagulation via Ets-1 Transcription Factor. Int J Mol Sci 2021; 23:244. [PMID: 35008670 PMCID: PMC8745726 DOI: 10.3390/ijms23010244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 12/20/2022] Open
Abstract
Scleroderma renal crisis (SRC) is an acute life-threatening manifestation of systemic sclerosis (SSc) caused by obliterative vasculopathy and thrombotic microangiopathy. Evidence suggests a pathogenic role of immunoglobulin G (IgG) targeting G-protein coupled receptors (GPCR). We therefore dissected SRC-associated vascular obliteration and investigated the specific effects of patient-derived IgG directed against angiotensin II type 1 (AT1R) and endothelin-1 type A receptors (ETAR) on downstream signaling events and endothelial cell proliferation. SRC-IgG triggered endothelial cell proliferation via activation of the mitogen-activated protein kinase (MAPK) pathway and subsequent activation of the E26 transformation-specific-1 transcription factor (Ets-1). Either AT1R or ETAR receptor inhibitors/shRNA abrogated endothelial proliferation, confirming receptor activation and Ets-1 signaling involvement. Binding of Ets-1 to the tissue factor (TF) promoter exclusively induced TF. In addition, TF inhibition prevented endothelial cell proliferation. Thus, our data revealed a thus far unknown link between SRC-IgG-induced intracellular signaling, endothelial cell proliferation and active coagulation in the context of obliterative vasculopathy and SRC. Patients' autoantibodies and their molecular effectors represent new therapeutic targets to address severe vascular complications in SSc.
Collapse
Affiliation(s)
- Rusan Catar
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Melanie Herse-Naether
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Nan Zhu
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200025, China
| | - Philine Wagner
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Oskar Wischnewski
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Angelika Kusch
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, 10117 Berlin, Germany
| | - Julian Kamhieh-Milz
- Department of Transfusion Medicine, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany;
| | - Andreas Eisenreich
- Department of Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.E.); (U.R.)
| | - Ursula Rauch
- Department of Cardiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (A.E.); (U.R.)
| | - Björn Hegner
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Vitanas Klinik für Geriatrie, 13435 Berlin, Germany
| | | | - Angela Kill
- Deutsches Rheuma-Forschungszentrum (DRFZ), A. Leibniz Institute, 10117 Berlin, Germany; (A.K.); (G.R.)
- Department of Rheumatology and Clinical Immunology, CCM, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Gabriela Riemekasten
- Deutsches Rheuma-Forschungszentrum (DRFZ), A. Leibniz Institute, 10117 Berlin, Germany; (A.K.); (G.R.)
- Department of Rheumatology and Clinical Immunology, CCM, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Members of the German Center for Lung Research (DZL), 23845 Borstel, Germany
| | - Gunnar Kleinau
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (G.K.); (P.S.)
| | - Patrick Scheerer
- Group Protein X-ray Crystallography and Signal Transduction, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (G.K.); (P.S.)
- DZHK (Deutsches Zentrum für Herz-Kreislauf Forschung), Partner Site Berlin, 13353 Berlin, Germany
| | - Duska Dragun
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, 10117 Berlin, Germany
| | - Aurelie Philippe
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (M.H.-N.); (N.Z.); (P.W.); (O.W.); (A.K.); (B.H.)
- Center for Cardiovascular Research, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health, Charité—Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, 10117 Berlin, Germany
| |
Collapse
|
30
|
Single-cell analysis of salt-induced hypertensive mouse aortae reveals cellular heterogeneity and state changes. Exp Mol Med 2021; 53:1866-1876. [PMID: 34862465 PMCID: PMC8741768 DOI: 10.1038/s12276-021-00704-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/26/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Elevated blood pressure caused by excessive salt intake is common and associated with cardiovascular diseases in most countries. However, the composition and responses of vascular cells in the progression of hypertension have not been systematically described. We performed single-cell RNA sequencing on the aortic arch from C57BL/6J mice fed a chow/high-salt diet. We identified 19 distinct cell populations representing 12 lineages, including smooth muscle cells (SMCs), fibroblasts, endothelial cells (ECs), B cells, and T cells. During the progression of hypertension, the proportion of three SMC subpopulations, two EC subpopulations, and T cells increased. In two EC clusters, the expression of reactive oxygen species-related enzymes, collagen and contractility genes was upregulated. Gene set enrichment analysis showed that three SMC subsets underwent endothelial-to-mesenchymal transition. We also constructed intercellular networks and found more frequent cell communication among aortic cells in hypertension and that some signaling pathways were activated during hypertension. Finally, joint public genome-wide association study data and our single-cell RNA-sequencing data showed the expression of hypertension susceptibility genes in ECs, SMCs, and fibroblasts and revealed 21 genes involved in the initiation and development of high-salt-induced hypertension. In conclusion, our data illustrate the transcriptional landscape of vascular cells in the aorta associated with hypertension and reveal dramatic changes in cell composition and intercellular communication during the progression of hypertension.
Collapse
|
31
|
Chen Y, Liu Y, Gao X. The Application of Single-Cell Technologies in Cardiovascular Research. Front Cell Dev Biol 2021; 9:751371. [PMID: 34708045 PMCID: PMC8542723 DOI: 10.3389/fcell.2021.751371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of deaths in the world. The intricacies of the cellular composition and tissue microenvironment in heart and vasculature complicate the dissection of molecular mechanisms of CVDs. Over the past decade, the rapid development of single-cell omics technologies generated vast quantities of information at various biological levels, which have shed light on the cellular and molecular dynamics in cardiovascular development, homeostasis and diseases. Here, we summarize the latest single-cell omics techniques, and show how they have facilitated our understanding of cardiovascular biology. We also briefly discuss the clinical value and future outlook of single-cell applications in the field.
Collapse
Affiliation(s)
- Yinan Chen
- Fuwai Hospital, Chinese Academy of Medical Sciences, Shenzhen, China.,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
| | - Yang Liu
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiang Gao
- Department of Vascular Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
32
|
Humphrey JD. Mechanisms of Vascular Remodeling in Hypertension. Am J Hypertens 2020; 34:432-441. [PMID: 33245319 PMCID: PMC8140657 DOI: 10.1093/ajh/hpaa195] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/09/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Hypertension is both a cause and a consequence of central artery stiffening, which in turn is an initiator and indicator of myriad disease conditions and thus all-cause mortality. Such stiffening results from a remodeling of the arterial wall that is driven by mechanical stimuli and mediated by inflammatory signals, which together lead to differential gene expression and concomitant changes in extracellular matrix composition and organization. This review focuses on biomechanical mechanisms by which central arteries remodel in hypertension within the context of homeostasis-what promotes it, what prevents it. It is suggested that the vasoactive capacity of the wall and inflammatory burden strongly influence the ability of homeostatic mechanisms to adapt the arterial wall to high blood pressure or not. Maladaptation, often reflected by inflammation-driven adventitial fibrosis, not just excessive intimal-medial thickening, significantly diminishes central artery function and disturbs hemodynamics, ultimately compromising end organ perfusion and thus driving the associated morbidity and mortality. It is thus suggested that there is a need for increased attention to controlling both smooth muscle phenotype and inflammation in hypertensive remodeling of central arteries, with future studies of the often adaptive response of medium-sized muscular arteries promising to provide additional guidance.
Collapse
Affiliation(s)
- Jay D Humphrey
- Department of Biomedical Engineering, Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA,Correspondence: Jay D. Humphrey ()
| |
Collapse
|
33
|
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
|