1
|
Hansen CJ, Rogers JH, Brown AJ, Boatwright N, Siricilla S, O'Brien CM, Panja S, Nichols CM, Devanathan K, Hardy BM, Does MD, Anderson AW, Paria BC, Mahadevan-Jansen A, Reese J, Herington JL. Regional differences in three-dimensional fiber organization, smooth muscle cell phenotype, and contractility in the pregnant mouse cervix. SCIENCE ADVANCES 2024; 10:eadr3530. [PMID: 39693423 DOI: 10.1126/sciadv.adr3530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024]
Abstract
The orientation and function of smooth muscle in the cervix may contribute to the important biomechanical properties that change during pregnancy. Thus, this study examined the three-dimensional structure, smooth muscle phenotype, and mechanical and contractile functions of the upper and lower cervix of nongravid (not pregnant) and gravid (pregnant) mice. In gravid cervix, we uncovered region-specific changes in the structure and organization of fiber tracts. We also detected a greater proportion of contractile smooth muscle cells (SMCs), but an equal proportion of synthetic SMCs, in the upper versus lower cervix. Furthermore, we revealed that the lower cervix had infrequent spontaneous contractions, distension had a minimal effect on contractility, and the upper cervix had forceful contractions in response to labor-inducing agents (oxytocin and prostaglandin E2). These findings identify regional differences in cervix contractility related to contractile SMC content and fiber organization, which could be targeted with diagnostic technologies and for therapeutic intervention.
Collapse
Affiliation(s)
- Christopher J Hansen
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Jackson H Rogers
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexus J Brown
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naoko Boatwright
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shajila Siricilla
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christine M O'Brien
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Sourav Panja
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cameron M Nichols
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kanchana Devanathan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville TN USA
| | - Benjamin M Hardy
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark D Does
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Adam W Anderson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bibhash C Paria
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anita Mahadevan-Jansen
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Biophotonics Center, Vanderbilt University, Nashville TN USA
- Department of Surgery, Neurological Surgery and Otolaryngology, Vanderbilt University Medical Center, Nashville, TN USA
| | - Jeff Reese
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Jennifer L Herington
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
2
|
Jacob AG, Moutsopoulos I, Petchey A, Kollyfas R, Knight-Schrijver VR, Mohorianu I, Sinha S, Smith CWJ. RNA binding protein with multiple splicing (RBPMS) promotes contractile phenotype splicing in human embryonic stem cell-derived vascular smooth muscle cells. Cardiovasc Res 2024; 120:2104-2116. [PMID: 39248180 PMCID: PMC11646123 DOI: 10.1093/cvr/cvae198] [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: 02/02/2024] [Revised: 06/12/2024] [Accepted: 07/14/2024] [Indexed: 09/10/2024] Open
Abstract
AIMS Differentiated vascular smooth muscle cells (VSMCs) express a unique network of mRNA isoforms via smooth muscle-specific alternative pre-mRNA splicing (SM-AS) in functionally critical genes, including those comprising the contractile machinery. We previously described RNA Binding Protein with Multiple Splicing (RBPMS) as a potent driver of differentiated SM-AS in the rat PAC1 VSMC cell line. What is unknown is how RBPMS affects VSMC phenotype and behaviour. Here, we aimed to dissect the role of RBPMS in SM-AS in human cells and determine the impact on VSMC phenotypic properties. METHODS AND RESULTS We used human embryonic stem cell-derived VSMCs (hESC-VSMCs) as our platform. hESC-VSMCs are inherently immature, and we found that they display only partially differentiated SM-AS patterns while RBPMS protein levels are low. We found that RBPMS over-expression induces SM-AS patterns in hESC-VSMCs akin to the contractile tissue VSMC splicing patterns. We present in silico and experimental findings that support RBPMS' splicing activity as mediated through direct binding and via functional cooperativity with splicing factor RBFOX2 on a significant subset of targets. We also demonstrate that RBPMS can alter the motility and the proliferative properties of hESC-VSMCs to mimic a more differentiated state. CONCLUSION Overall, this study emphasizes a critical role for RBPMS in establishing the contractile phenotype splicing programme of human VSMCs.
Collapse
Affiliation(s)
- Aishwarya G Jacob
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | | | - Alex Petchey
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Rafael Kollyfas
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | | | - Irina Mohorianu
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | - Sanjay Sinha
- MRC-Wellcome Cambridge Stem Cell Institute, Cambridge CB2 0AW, UK
| | | |
Collapse
|
3
|
Maegdefessel L, Fasolo F. Long Noncoding RNA Function in Smooth Muscle Cell Plasticity and Atherosclerosis. Arterioscler Thromb Vasc Biol 2024. [PMID: 39633574 DOI: 10.1161/atvbaha.124.320393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
In the healthy mature artery, vascular cells, including endothelial cells, smooth muscle cells (SMCs), and fibroblasts are organized in different layers, performing specific functions. SMCs located in the media are in a differentiated state and exhibit a contractile phenotype. However, in response to vascular injury within the intima, stimuli from activated endothelial cells and recruited inflammatory cells reach SMCs and induce a series of remodeling events in them, known as phenotypic switching. Indeed, SMCs retain a certain degree of plasticity and are able to transdifferentiate into other cell types that are crucial for both the formation and development of atherosclerotic lesions. Because of their highly cell-specific expression profiles and their widely recognized contribution to physiological and disease-related biological processes, long noncoding RNAs have received increasing attention in atherosclerosis research. Dynamic fluctuations in their expression have been implicated in the regulation of SMC identity. Sophisticated technologies are now available to allow researchers to access single-cell transcriptomes and study long noncoding RNA function with unprecedented precision. Here, we discuss the state of the art of long noncoding RNAs regulation of SMC phenotypic switching, describing the methodologies used to approach this issue and evaluating the therapeutic perspectives of exploiting long noncoding RNAs as targets in atherosclerosis.
Collapse
Affiliation(s)
- Lars Maegdefessel
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany (L.M., F.F.)
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Berlin, Germany (L.M., F.F.)
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden (L.M.)
| | - Francesca Fasolo
- Institute of Molecular Vascular Medicine, Klinikum rechts der Isar, Technical University Munich, Germany (L.M., F.F.)
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Berlin, Germany (L.M., F.F.)
| |
Collapse
|
4
|
Bramel EE, Espinoza Camejo WA, Creamer TJ, Restrepo L, Saqib M, Bagirzadeh R, Zeng A, Mitchell JT, Stein-O'Brien GL, Pedroza AJ, Fischbein MP, Dietz HC, Gallo MacFarlane E. Intrinsic GATA4 expression sensitizes the aortic root to dilation in a Loeys-Dietz syndrome mouse model. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1468-1481. [PMID: 39567770 PMCID: PMC11634776 DOI: 10.1038/s44161-024-00562-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024]
Abstract
Loeys-Dietz syndrome (LDS) is a connective tissue disorder caused by mutations that decrease transforming growth factor-β signaling. LDS-causing mutations increase the risk of aneurysm throughout the arterial tree, yet the aortic root is a site of heightened susceptibility. Here we investigate the heterogeneity of vascular smooth muscle cells (VSMCs) in the aorta of Tgfbr1M318R/+ LDS mice by single-cell transcriptomics to identify molecular determinants of this vulnerability. Reduced expression of components of the extracellular matrix-receptor apparatus and upregulation of stress and inflammatory pathways were observed in all LDS VSMCs. However, regardless of genotype, a subset of Gata4-expressing VSMCs predominantly located in the aortic root intrinsically displayed a less differentiated, proinflammatory profile. A similar population was also identified among aortic VSMCs in a human single-cell RNA sequencing dataset. Postnatal VSMC-specific Gata4 deletion reduced aortic root dilation in LDS mice, suggesting that this factor sensitizes the aortic root to the effects of impaired transforming growth factor-β signaling.
Collapse
MESH Headings
- Animals
- Loeys-Dietz Syndrome/genetics
- Loeys-Dietz Syndrome/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Disease Models, Animal
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- GATA4 Transcription Factor/genetics
- GATA4 Transcription Factor/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Receptor, Transforming Growth Factor-beta Type I/metabolism
- Humans
- Signal Transduction
- Dilatation, Pathologic/genetics
- Male
- Aorta/metabolism
- Aorta/pathology
- Single-Cell Analysis
- Mice
- Aortic Aneurysm/genetics
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Mice, Inbred C57BL
- Transcriptome
- Mice, Knockout
Collapse
Affiliation(s)
- Emily E Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wendy A Espinoza Camejo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tyler J Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Leda Restrepo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Muzna Saqib
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rustam Bagirzadeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony Zeng
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacob T Mitchell
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Genevieve L Stein-O'Brien
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Albert J Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Harry C Dietz
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
5
|
Yang WY, Ben Issa M, Saaoud F, Xu K, Shao Y, Lu Y, Dornas W, Cueto R, Jiang X, Wang H, Yang X. Perspective: Pathological transdifferentiation-a novel therapeutic target for cardiovascular diseases and chronic inflammation. Front Cardiovasc Med 2024; 11:1500775. [PMID: 39660114 PMCID: PMC11628510 DOI: 10.3389/fcvm.2024.1500775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Pathological transdifferentiation, where differentiated cells aberrantly transform into other cell types that exacerbate disease rather than promote healing, represents a novel and significant concept. This perspective discusses its role and potential targeting in cardiovascular diseases and chronic inflammation. Current therapies mainly focus on mitigating early inflammatory response through proinflammatory cytokines and pathways targeting, including corticosteroids, TNF-α inhibitors, IL-1β monoclonal antibodies and blockers, IL-6 blockers, and nonsteroidal anti-inflammatory drugs (NSAIDs), along with modulating innate immune memory (trained immunity). However, these approaches often fail to address long-term tissue damage and functional regeneration. For instance, fibroblasts can transdifferentiate into myofibroblasts in cardiac fibrosis, and endothelial cells may undergo endothelial to mesenchymal transition (EndMT) in vascular remodeling, resulting in fibrosis and impaired tissue function. Targeting pathological transdifferentiation represents a promising therapeutic avenue by focusing on key signaling pathways that drive these aberrant cellular phenotypic and transcriptomic transitions. This approach seeks to inhibit these pathways or modulate cellular plasticity to promote effective tissue regeneration and prevent fibrosis. Such strategies have the potential to address inflammation, cell death, and the resulting tissue damage, providing a more comprehensive and sustainable treatment solution. Future research should focus on understanding the mechanisms behind pathological transdifferentiation, identifying relevant biomarkers and master regulators, and developing novel therapies through preclinical and clinical trials. Integrating these new therapies with existing anti-inflammatory treatments could enhance efficacy and improve patient outcomes. Highlighting pathological transdifferentiation as a therapeutic target could transform treatment paradigms, leading to better management and functional recovery of cardiovascular tissues in diseases and chronic inflammation.
Collapse
Affiliation(s)
- William Y. Yang
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Mohammed Ben Issa
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Fatma Saaoud
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Keman Xu
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yifan Lu
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Waleska Dornas
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Ramon Cueto
- Department of Cardiovascular Sciences, Metabolic Disease Research and Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
- Department of Cardiovascular Sciences, Metabolic Disease Research and Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Department of Cardiovascular Sciences, Metabolic Disease Research and Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences, Lemole Center for Integrated Lymphatics and Vascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
- Department of Cardiovascular Sciences, Metabolic Disease Research and Thrombosis Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
6
|
Qi RQ, Chen YF, Cheng J, Song JW, Chen YH, Wang SY, Liu Y, Yan KX, Liu XY, Li J, Zhong JC. Elabela alleviates cuproptosis and vascular calcification in vitaminD3- overloaded mice via regulation of the PPAR-γ /FDX1 signaling. Mol Med 2024; 30:223. [PMID: 39567863 PMCID: PMC11577739 DOI: 10.1186/s10020-024-00997-3] [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: 08/30/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Vascular calcification is a crucial pathophysiological process associated with age-related cardiovascular diseases. Elabela, a recently identified peptide, has emerged as a significant player in the regulation of cardiovascular function and homeostasis. However, the effects and underlying mechanisms of Elabela on age-related vascular calcification remain largely unexplored. METHODS In-vivo vascular calcifications of C57BL/6J mice (8-week-old) and young (8-week-old) or aged (72-week-old) SD rats were injected with vitamin D3 (VitD3) or saline, respectively. Furthermore, the VitD3-overloaded mice received Elabela (1 mg/kg/d), peroxisome proliferators-activated receptor-γ (PPAR-γ) activator Rosiglitazone (5 mg/kg/d) or copper-ionophore Elesclomol (20 mg/kg/d), respectively. As for in-vitro studies, primary rat vascular smooth muscle cells (VSMCs) were isolated from aortas and cultured for explore the role and underlying mechanism of Elabela in vascular calcification. RESULTS There were marked increases in FDX1 and Slc31a1 levels in both aortas and VSMCs during vascular calcification, coinciding with a rise in copper levels and a decrease in Elabela levels. Alizarin red and von-Kossa staining indicated that the administration of Elabela effectively hindered the progression of vascular cuproptosis and arterial calcification in VitD3-overloaded mice and rat arterial rings models. Moreover, Elabela significantly suppressed osteogenic differentiation and calcium deposition in VSMCs and strikingly reversed high phosphate-induced augmentation of FDX1 expression, DLAT aggregation as well as intracellular copper ion levels. More importantly, Elabela exhibited remarkable abilities to prevent mitochondrial dysfunctions in primary rat VSMCs by maintaining mitochondrial membrane potential, inhibiting mitochondrial division, reducing mitochondrial ROS production and increasing ATP levels. Interestingly, Elabela mitigated cellular senescence and production of pro-inflammatory cytokines including IL-1α, IL-1β, IL-6, IL-18 and TNF-α, respectively. Furthermore, Elabela upregulated the protein levels of PPAR-γ in VitD3-overloaded mice. Administrating PPAR-γ inhibitor GW9662 or blocking the efflux of intracellular copper abolished the protective effect of Elabela on vascular calcification by enhancing levels of FDX1, Slc31a1, Runx2, and BMP2. CONCLUSION Elabela plays a crucial role in protecting against vascular cuproptosis and arterial calcification by activating the PPAR-γ /FDX1 signaling. Elabela supplementation and cuproptosis suppression serve as effective therapeutic approaches for managing vascular calcification and related cardiovascular disorders.
Collapse
Affiliation(s)
- Rui-Qiang Qi
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yu-Fei Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing Cheng
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jia-Wei Song
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yi-Hang Chen
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Si-Yuan Wang
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Ying Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Kai-Xin Yan
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Xiao-Yan Liu
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Jing Li
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China
| | - Jiu-Chang Zhong
- Heart Center and Beijing Key Laboratory of Hypertension, Beijing Chaoyang Hospital and Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing, 100020, China.
- Department of Cardiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
7
|
Song J, Cao C, Wang Z, Li H, Yang L, Kang J, Meng H, Li L, Liu J. Mechanistic insights into the regression of atherosclerotic plaques. Front Physiol 2024; 15:1473709. [PMID: 39628943 PMCID: PMC11611857 DOI: 10.3389/fphys.2024.1473709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
Atherosclerosis is a major contributor to cardiovascular diseases and mortality globally. The progression of atherosclerotic disease results in the expansion of plaques and the development of necrotic cores. Subsequent plaque rupture can lead to thrombosis, occluding blood vessels, and end-organ ischemia with consequential ischemic injury. Atherosclerotic plaques are formed by the accumulation of lipid particles overloaded in the subendothelial layer of blood vessels. Abnormally elevated blood lipid levels and impaired endothelial function are the initial factors leading to atherosclerosis. The atherosclerosis research has never been interrupted, and the previous view was that the pathogenesis of atherosclerosis is an irreversible and chronic process. However, recent studies have found that the progression of atherosclerosis can be halted when patients' blood lipid levels are reversed to normal or lower. A large number of studies indicates that it can inhibit the progression of atherosclerosis lesions and promote the regression of atherosclerotic plaques and necrotic cores by lowering blood lipid levels, improving the repair ability of vascular endothelial cells, promoting the reverse cholesterol transport in plaque foam cells and enhancing the ability of macrophages to phagocytize and clear the necrotic core of plaque. This article reviews the progress of research on the mechanism of atherosclerotic plaque regression. Our goal is to provide guidance for developing better therapeutic approaches to atherosclerosis by reviewing and analyzing the latest scientific findings.
Collapse
Affiliation(s)
- Jianshu Song
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Ce Cao
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Ziyan Wang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Haoran Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Lili Yang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jing Kang
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Hongxu Meng
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Lei Li
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Jianxun Liu
- National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
8
|
Li K, Zhang C, Wang LX, Wang X, Wang R. KLF4's role in regulating nitric oxide production and promoting microvascular formation following ischemic stroke. Nitric Oxide 2024; 154:86-104. [PMID: 39557151 DOI: 10.1016/j.niox.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
This study examines KLF4's role in endothelial cells (ECs), emphasizing its effects on nitric oxide (NO) production, microvascular formation, and oxidative stress regulation following ischemic stroke. Through high-throughput sequencing, we identified eight cell subpopulations in carotid artery tissues post-stroke, with KLF4 notably elevated in ECs. KLF4 overexpression in ECs promoted NO synthesis, enhanced endothelial tube formation, mitigated oxidative stress, and improved smooth muscle cells (SMCs) function, collectively boosting blood flow in ischemic regions. These findings highlight KLF4 as pivotal in vascular regeneration and oxidative stress reduction, positioning it as a promising target for cardiovascular and cerebrovascular therapies.
Collapse
Affiliation(s)
- Kuo Li
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China.
| | - Chuansuo Zhang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Li Xuan Wang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Xiaoxuan Wang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Ruyue Wang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| |
Collapse
|
9
|
Deng W, Huang S, Yu L, Gao B, Pan Y, Wang X, Li L. HIF-1α knockdown attenuates phenotypic transformation and oxidative stress induced by high salt in human aortic vascular smooth muscle cells. Sci Rep 2024; 14:28100. [PMID: 39543255 PMCID: PMC11564746 DOI: 10.1038/s41598-024-79892-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/13/2024] [Indexed: 11/17/2024] Open
Abstract
Increased dietary salt intake is a well-established risk factor for hypertension and related cardiovascular diseases, involving complex vascular remodeling processes. However, the specific role of hypoxia-inducible factor-1α (HIF-1α) in vascular pathophysiology under high-salt conditions remains poorly understood. This study investigates the role of HIF-1α in high-salt-induced vascular remodeling using human aortic vascular smooth muscle cells (HA-VSMCs) cultured in vitro. HA-VSMCs were divided into three groups: high-salt with HIF-1α knockdown (shHIF-1α + HS), negative control (shcontrol), and high-salt (HS). Cell viability, migration, gene expression, and protein levels were evaluated. High-salt conditions significantly increased mRNA expression of α-smooth muscle actin (α-SMA), smooth muscle protein 22 (SM22), angiotensin II type 1 receptor (AT1R), collagen I, and collagen III (p < 0.0001). HIF-1α knockdown partially attenuated these increases, particularly for α-SMA, SM22, and AT1R (p < 0.01). At the protein level, high-salt exposure markedly elevated expression of collagen III, HIF-1α, osteopontin (OPN), and angiotensin II (Ang II) (p < 0.0001). HIF-1α knockdown significantly reduced the high-salt-induced increases in collagen III and HIF-1α protein levels (p < 0.001) but had a limited effect on OPN and Ang II upregulation. Interestingly, SM22 protein expression was significantly decreased under high-salt conditions (p < 0.0001), an effect partially reversed by HIF-1α knockdown (p < 0.0001). These findings demonstrate that high-salt conditions induce complex changes in gene and protein expression in HA-VSMCs, with HIF-1α playing a crucial role in mediating many of these alterations. The study highlights the differential effects of HIF-1α on various markers of vascular remodeling and suggests that HIF-1α may be a potential therapeutic target for mitigating salt-induced vascular pathology. Further research is warranted to elucidate the mechanisms underlying the HIF-1α-dependent and -independent effects observed in this study.
Collapse
MESH Headings
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Oxidative Stress/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Aorta/metabolism
- Aorta/cytology
- Gene Knockdown Techniques
- Sodium Chloride, Dietary/adverse effects
- Actins/metabolism
- Phenotype
- Cell Movement/drug effects
- Muscle Proteins/metabolism
- Muscle Proteins/genetics
- Microfilament Proteins/metabolism
- Microfilament Proteins/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Vascular Remodeling/drug effects
- Cell Survival/drug effects
- Osteopontin/metabolism
- Osteopontin/genetics
- Cells, Cultured
Collapse
Affiliation(s)
- Wenbin Deng
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Shiqiong Huang
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Lisha Yu
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Bo Gao
- Department of Pathology, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Yun Pan
- Department of Pathology, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Xue Wang
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China
| | - Lihua Li
- Department of Geriatrics, The First Affiliated Hospital of Dali University, Dali, 671000, Yunnan, China.
- Department of Gerontology, The First Affiliated Hospital of Dali University, Jiashibo Road 32, Dali, 671000, China.
| |
Collapse
|
10
|
Gastanadui MG, Margaroli C, Litovsky S, Richter RP, Wang D, Xing D, Wells JM, Gaggar A, Nanda V, Patel RP, Payne GA. Spatial Transcriptomic Approach to Understanding Coronary Atherosclerotic Plaque Stability. Arterioscler Thromb Vasc Biol 2024; 44:e264-e276. [PMID: 39234691 PMCID: PMC11499036 DOI: 10.1161/atvbaha.123.320330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 08/08/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Coronary atherosclerotic plaques susceptible to acute coronary syndrome have traditionally been characterized by their surrounding cellular architecture. However, with the advent of intravascular imaging, novel mechanisms of coronary thrombosis have emerged, challenging our contemporary understanding of acute coronary syndrome. These intriguing findings underscore the necessity for a precise molecular definition of plaque stability. Considering this, our study aimed to investigate the vascular microenvironment in patients with stable and unstable plaques using spatial transcriptomics. METHODS Autopsy-derived coronary arteries were preserved and categorized by plaque stability (n=5 patients per group). We utilized the GeoMx spatial profiling platform and Whole Transcriptome Atlas to link crucial histological morphology markers in coronary lesions with differential gene expression in specific regions of interest, thereby mapping the vascular transcriptome. This innovative approach allowed us to conduct cell morphological and spatially resolved transcriptional profiling of atherosclerotic plaques while preserving crucial intercellular signaling. RESULTS We observed intriguing spatial and cell-specific transcriptional patterns in stable and unstable atherosclerotic plaques, showcasing regional variations within the intima and media. These regions exhibited differential expression of proinflammatory molecules (eg, IFN-γ [interferon-γ], MHC [major histocompatibility complex] class II, proinflammatory cytokines) and prothrombotic signaling pathways. By using lineage tracing through spatial deconvolution of intimal CD68+ (cluster of differentiation 68) cells, we characterized unique, intraplaque subpopulations originating from endothelial, smooth muscle, and myeloid lineages with distinct regional locations associated with plaque instability. In addition, unique transcriptional signatures were observed in vascular smooth muscle and CD68+ cells among plaques exhibiting coronary calcification. CONCLUSIONS Our study illuminates distinct cell-specific and regional transcriptional alterations present in unstable plaques. Furthermore, we characterize spatially resolved, in situ evidence supporting cellular transdifferentiation and intraplaque plasticity as significant contributors to plaque instability in human coronary atherosclerosis. Our results provide a powerful resource for the identification of novel mediators of acute coronary syndrome, opening new avenues for preventative and therapeutic treatments.
Collapse
Affiliation(s)
- Maria G Gastanadui
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Camilla Margaroli
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Silvio Litovsky
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert P. Richter
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, Division of Pediatric Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dezhi Wang
- Department of Pathology, Pathology Core Research Laboratory, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dongqi Xing
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J. Michael Wells
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
| | - Amit Gaggar
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Medicine, Division of Pulmonary, Allergy & Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Vivek Nanda
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rakesh P. Patel
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pathology, Division of Molecular & Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gregory A. Payne
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, USA
- Cardiopulmonary Research Program, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, USA
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, USA
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL
| |
Collapse
|
11
|
Wang Y, Thottappillil N, Gomez-Salazar M, Tower RJ, Qin Q, Del Rosario Alvia IC, Xu M, Cherief M, Cheng R, Archer M, Arondekar S, Reddy S, Broderick K, Péault B, James AW. Integrated transcriptomics of human blood vessels defines a spatially controlled niche for early mesenchymal progenitor cells. Dev Cell 2024; 59:2687-2703.e6. [PMID: 39025061 PMCID: PMC11496018 DOI: 10.1016/j.devcel.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/28/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024]
Abstract
Human blood vessel walls show concentric layers, with the outermost tunica adventitia harboring mesenchymal progenitor cells. These progenitor cells maintain vessel homeostasis and provide a robust cell source for cell-based therapies. However, human adventitial stem cell niche has not been studied in detail. Here, using spatial and single-cell transcriptomics, we characterized the phenotype, potential, and microanatomic distribution of human perivascular progenitors. Initially, spatial transcriptomics identified heterogeneity between perivascular layers of arteries and veins and delineated the tunica adventitia into inner and outer layers. From this spatial atlas, we inferred a hierarchy of mesenchymal progenitors dictated by a more primitive cell with a high surface expression of CD201 (PROCR). When isolated from humans and mice, CD201Low expression typified a mesodermal committed subset with higher osteogenesis and less proliferation than CD201High cells, with a downstream effect on canonical Wnt signaling through DACT2. CD201Low cells also displayed high translational potential for bone tissue generation.
Collapse
Affiliation(s)
- Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | - Robert J Tower
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Mingxin Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Masnsen Cherief
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Ray Cheng
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mary Archer
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Shreya Arondekar
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sashank Reddy
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Kristen Broderick
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Bruno Péault
- Department of Orthopedic Surgery and Orthopedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
12
|
Wolfson SM, Beigel K, Anderson SE, Deal B, Weiner M, Lee SH, Taylor D, Heo SC, Heuckeroth RO, Hashmi SK. Rapid cyclic stretching induces a synthetic, proinflammatory phenotype in cultured human intestinal smooth muscle, with the potential to alter signaling to adjacent bowel cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.12.617767. [PMID: 39464046 PMCID: PMC11507745 DOI: 10.1101/2024.10.12.617767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Background and Aims Bowel smooth muscle experiences mechanical stress constantly during normal function, and pathologic mechanical stressors in disease states. We tested the hypothesis that pathologic mechanical stress could alter transcription to induce smooth muscle phenotypic class switching. Methods Primary human intestinal smooth muscle cells (HISMCs), seeded on electrospun aligned poly-ε-caprolactone nano-fibrous scaffolds, were subjected to pathologic, high frequency (1 Hz) uniaxial 3% cyclic stretch (loaded) or kept unloaded in culture for 6 hours. Total RNA sequencing, qRT-PCR, and quantitative immunohistochemistry defined loading-induced changes in gene expression. NicheNet predicted how differentially expressed genes might impact HISMCs and other bowel cells. Results Loading induced differential expression of 4537 genes in HISMCs. Loaded HISMCs had a less contractile phenotype, with increased expression of synthetic SMC genes, proinflammatory cytokines, and altered expression of axon guidance molecules, growth factors and morphogens. Many differentially expressed genes encode secreted ligands that could act cell-autonomously on smooth muscle and on other cells in the bowel wall. Discussion HISMCs demonstrate remarkably rapid phenotypic plasticity in response to mechanical stress that may convert contractile HISMCs into proliferative, fibroblast-like cells or proinflammatory cells. These mechanical stress-induced changes in HISMC gene expression may be relevant for human bowel disease.
Collapse
Affiliation(s)
- Sharon M. Wolfson
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Katherine Beigel
- The Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sierra E. Anderson
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Brooke Deal
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Molly Weiner
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | | | - Deanne Taylor
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Department of Biomedical and Health Informatics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Chin Heo
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Bioengineering, The University of Pennsylvania School of Engineering and Applied Science, 220 S 33rd St, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Robert O. Heuckeroth
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sohaib K. Hashmi
- The Children’s Hospital of Philadelphia Research Institute and the Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Bioengineering, The University of Pennsylvania School of Engineering and Applied Science, 220 S 33rd St, Philadelphia, PA 19104, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Hospital of the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
13
|
Qian Y, Xiong S, Li L, Sun Z, Zhang L, Yuan W, Cai H, Feng G, Wang X, Yao H, Gao Y, Guo L, Wang Z. Spatial multiomics atlas reveals smooth muscle phenotypic transformation and metabolic reprogramming in diabetic macroangiopathy. Cardiovasc Diabetol 2024; 23:358. [PMID: 39395983 PMCID: PMC11471023 DOI: 10.1186/s12933-024-02458-x] [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: 07/17/2024] [Accepted: 09/25/2024] [Indexed: 10/14/2024] Open
Abstract
BACKGROUND Diabetic macroangiopathy has been the main cause of death and disability in diabetic patients. The mechanisms underlying smooth muscle cell transformation and metabolic reprogramming other than abnormal glucose and lipid metabolism remain to be further explored. METHOD Single-cell transcriptome, spatial transcriptome and spatial metabolome sequencing were performed on anterior tibial artery from 11 diabetic patients with amputation. Multi-omics integration, cell communication analysis, time series analysis, network analysis, enrichment analysis, and gene expression analysis were performed to elucidate the potential molecular features. RESULT We constructed a spatial multiomics map of diabetic blood vessels based on multiomics integration, indicating single-cell and spatial landscape of transcriptome and spatial landscape of metabolome. At the same time, the characteristics of cell composition and biological function of calcified regions were obtained by integrating spatial omics and single cell omics. On this basis, our study provides favorable evidence for the cellular fate of smooth muscle cells, which can be transformed into pro-inflammatory chemotactic smooth muscle cells, macrophage-like smooth muscle cells/foam-like smooth muscle cells, and fibroblast/chondroblast smooth muscle cells in the anterior tibial artery of diabetic patients. The smooth muscle cell phenotypic transformation is driven by transcription factors net including KDM5B, DDIT3, etc. In addition, in order to focus on metabolic reprogramming apart from abnormal glucose and lipid metabolism, we constructed a metabolic network of diabetic vascular activation, and found that HNMT and CYP27A1 participate in diabetic vascular metabolic reprogramming by combining public data. CONCLUSION This study constructs the spatial gene-metabolism map of the whole anterior tibial artery for the first time and reveals the characteristics of vascular calcification, the phenotypic transformation trend of SMCs, and the transcriptional driving network of SMCs phenotypic transformation of diabetic macrovascular disease. In the perspective of combining the transcriptome and metabolome, the study demonstrates the activated metabolic pathways in diabetic blood vessels and the key genes involved in diabetic metabolic reprogramming.
Collapse
Affiliation(s)
- Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 212001, China
| | - Shizheng Xiong
- State Key Laboratory of Organic Electronics and Information Displays and Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 212001, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 212001, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 212001, China
| | - Honghua Cai
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guoquan Feng
- Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiaoguang Wang
- Department of Joint Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Haipeng Yao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 212001, China
| | - Yun Gao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays and Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing, 210023, China.
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 212001, China.
| |
Collapse
|
14
|
Huang A, Rao J, Feng X, Li X, Xu T, Yao L. Breaking new ground: Unraveling the USP1/ID3/E12/P21 axis in vascular calcification. Transl Res 2024; 276:1-20. [PMID: 39326697 DOI: 10.1016/j.trsl.2024.09.002] [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: 05/05/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Vascular calcification (VC) poses significant challenges in cardiovascular health. This study employs single-cell transcriptome sequencing to dissect cellular dynamics in this process. We identify distinct cell subgroups, notably in vascular smooth muscle cells (VSMCs), and observe differences between calcified atherosclerotic cores and adjacent regions. Further exploration reveals ID3 as a key gene regulating VSMC function. In vitro experiments demonstrate ID3's interaction with USP1 and E12, modulating cell proliferation and osteogenic differentiation. Animal models confirm the critical role of the USP1/ID3/E12/P21 axis in VC. This study sheds light on a novel regulatory mechanism, offering potential therapeutic targets.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, PR China
| | - Jianyun Rao
- Outpatient Management Office, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, PR China
| | - Xin Feng
- Department of Nephrology, Liaoning electric power central hospital, Shenyang 110000, PR China
| | - Xingru Li
- Department of Nephrology, Liaoning electric power central hospital, Shenyang 110000, PR China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, PR China.
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang 110000, PR China.
| |
Collapse
|
15
|
van Merrienboer TAR, Rombouts KB, Bogunovic N, Mieremet A, Meekel JP, Balm R, de Waard V, Yeung KK. Metformin Improves the Function of Abdominal Aortic Aneurysm Patient-Derived Aortic Smooth Muscle Cells. Eur J Vasc Endovasc Surg 2024:S1078-5884(24)00814-1. [PMID: 39321955 DOI: 10.1016/j.ejvs.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 08/06/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE Type 2 diabetes mellitus (T2DM) is a cardiovascular risk factor. Paradoxically, a decreased risk of abdominal aortic aneurysm (AAA) presence and growth rate is described among patients with T2DM, associated with metformin use. This study aimed to investigate the effect of metformin on AAA patient-derived aortic smooth muscle cell (SMC) function. METHODS Aortic biopsies were obtained from patients with AAA (n = 21) and controls (n = 17) during surgery. The SMCs of non-pathological aortic controls, non-diabetic patients with AAA, and diabetic patients with AAA were cultured from explants and treated with or without metformin. The SMC contractility was measured upon ionomycin stimulation, as well as metabolic activity, proliferation, and migration. mRNA and protein expression of markers for contraction, metabolic activity, proliferation, and inflammation were measured. RESULTS mRNA expression of KLF4 and GYS1, genes involved in metabolic activity, differed between SMCs from non-diabetic and diabetic patients with AAA before metformin stimulation (p < .041). However, the effect of metformin on the various SMC functions was similar between non-diabetic and diabetic patients with AAA. Upon stimulation, metformin increased the contractility of AAA patient SMCs (p = .001). mRNA expression of smoothelin, a marker for the contractile phenotype, increased in SMCs of patients with AAA after treatment with metformin (p = .006). An increase in metabolic activity (p < .001) and a decrease in proliferation (p < .001) and migration were found in the SMCs of controls and patients with AAA with metformin. Increased mRNA expression of PPARγ, a nuclear receptor involved in mitochondrial biogenesis (p < .009), and a decrease in gene expression of Ki-67, a marker for proliferation (p < .005), were observed. Gene expression of inflammation markers MCP-1 and IL-6, and protein expression of NF-κB p65 decreased after treatment with metformin in patients with AAA. CONCLUSION This study found that metformin increases contractility and metabolic activity, and reduces proliferation, migration, and inflammation in aortic SMCs in vitro.
Collapse
Affiliation(s)
- Tara A R van Merrienboer
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands.
| | - Karlijn B Rombouts
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| | - Natalija Bogunovic
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands
| | - Arnout Mieremet
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| | - Jorn P Meekel
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands
| | - Ron Balm
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands
| | - Vivian de Waard
- Amsterdam UMC location University of Amsterdam, Medical Biochemistry, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| | - Kak K Yeung
- Amsterdam UMC location University of Amsterdam, Surgery, Amsterdam, the Netherlands; Amsterdam UMC location Vrije Universiteit Amsterdam, Physiology, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischaemic Syndromes, Amsterdam, the Netherlands
| |
Collapse
|
16
|
Stankovics L, Ungvari A, Fekete M, Nyul-Toth A, Mukli P, Patai R, Csik B, Gulej R, Conley S, Csiszar A, Toth P. The vasoprotective role of IGF-1 signaling in the cerebral microcirculation: prevention of cerebral microhemorrhages in aging. GeroScience 2024:10.1007/s11357-024-01343-5. [PMID: 39271571 DOI: 10.1007/s11357-024-01343-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Aging is closely associated with various cerebrovascular pathologies that significantly impact brain function, with cerebral small vessel disease (CSVD) being a major contributor to cognitive decline in the elderly. Consequences of CSVD include cerebral microhemorrhages (CMH), which are small intracerebral bleeds resulting from the rupture of microvessels. CMHs are prevalent in aging populations, affecting approximately 50% of individuals over 80, and are linked to increased risks of vascular cognitive impairment and dementia (VCID). Hypertension is a primary risk factor for CMHs. Vascular smooth muscle cells (VSMCs) adapt to hypertension by undergoing hypertrophy and producing extracellular matrix (ECM) components, which reinforce vessel walls. Myogenic autoregulation, which involves pressure-induced constriction, helps prevent excessive pressure from damaging the vulnerable microvasculature. However, aging impairs these adaptive mechanisms, weakening vessel walls and increasing susceptibility to damage. Insulin-like Growth Factor 1 (IGF-1) is crucial for vascular health, promoting VSMC hypertrophy, ECM production, and maintaining normal myogenic protection. IGF-1 also prevents microvascular senescence, reduces reactive oxygen species (ROS) production, and regulates matrix metalloproteinase (MMP) activity, which is vital for ECM remodeling and stabilization. IGF-1 deficiency, common in aging, compromises these protective mechanisms, increasing the risk of CMHs. This review explores the vasoprotective role of IGF-1 signaling in the cerebral microcirculation and its implications for preventing hypertension-induced CMHs in aging. Understanding and addressing the decline in IGF-1 signaling with age are crucial for maintaining cerebrovascular health and preventing hypertension-related vascular injuries in the aging population.
Collapse
Affiliation(s)
- Levente Stankovics
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Adam Nyul-Toth
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Roland Patai
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Boglarka Csik
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rafal Gulej
- International Training Program in Geroscience, Doctoral College-Health Sciences Program/ Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Toth
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
- Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
17
|
Wu Y, Chen Z, Zheng Z, Li X, Shu J, Mao R, An J, Fan S, Luo R, Guo Y, Xu W, Liang M, Huang K, Wang C. Tudor-SN exacerbates pathological vascular remodeling by promoting the polyubiquitination of PTEN via NEDD4-1. J Biomed Sci 2024; 31:88. [PMID: 39237902 PMCID: PMC11378411 DOI: 10.1186/s12929-024-01076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Dysregulation of vascular homeostasis can induce cardiovascular diseases and increase global mortality rates. Although lineage tracing studies have confirmed the pivotal role of modulated vascular smooth muscle cells (VSMCs) in the progression of pathological vascular remodeling, the underlying mechanisms are still unclear. METHODS The expression of Tudor-SN was determined in VSMCs of artery stenosis, PDGF-BB-treated VSMCs and atherosclerotic plaque. Loss- and gain-of-function approaches were used to explore the role of Tudor-SN in the modulation of VSMCs phenotype both in vivo and in vitro. RESULTS In this study, we demonstrate that Tudor-SN expression is significantly elevated in injury-induced arteries, atherosclerotic plaques, and PDGF-BB-stimulated VSMCs. Tudor-SN deficiency attenuates, but overexpression aggravates the synthetic phenotypic switching of VSMCs and pathological vascular remodeling. Loss of Tudor-SN also reduces atherosclerotic plaque formation and increases plaque stability. Mechanistically, PTEN, the major regulator of the MAPK and PI3K-AKT signaling pathways, plays a vital role in Tudor-SN-mediated regulation on proliferation and migration of VSMCs. Tudor-SN facilitates the polyubiquitination and degradation of PTEN via NEDD4-1, thus exacerbating vascular remodeling under pathological conditions. BpV (HOpic), a specific inhibitor of PTEN, not only counteracts the protective effect of Tudor-SN deficiency on proliferation and migration of VSMCs, but also abrogates the negative effect of carotid artery injury-induced vascular remodeling in mice. CONCLUSIONS Our findings reveal that Tudor-SN deficiency significantly ameliorated pathological vascular remodeling by reducing NEDD4-1-dependent PTEN polyubiquitination, suggesting that Tudor-SN may be a novel target for preventing vascular diseases.
Collapse
Affiliation(s)
- Yichen Wu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China
| | - Zilong Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Zhe Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xiaoguang Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jiangcheng Shu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ruiqi Mao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Jie An
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Siyuan Fan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Ruijie Luo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Yi Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
| | - Wenjing Xu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China
| | - Minglu Liang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China
| | - Kai Huang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| | - Cheng Wang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, China.
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Ave, Wuhan, 430022, Hubei, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, China.
- Hubei Clinical Research Center for Metabolic and Cardiovascular Disease, Wuhan, China.
| |
Collapse
|
18
|
Jia K, Luo X, Yi J, Zhang C. Hormonal influence: unraveling the impact of sex hormones on vascular smooth muscle cells. Biol Res 2024; 57:61. [PMID: 39227995 PMCID: PMC11373308 DOI: 10.1186/s40659-024-00542-w] [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: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Sex hormones play a pivotal role as endocrine hormones that exert profound effects on the biological characteristics and vascular function of vascular smooth muscle cells (VSMCs). By modulating intracellular signaling pathways, activating nuclear receptors, and regulating gene expression, sex hormones intricately influence the morphology, function, and physiological state of VSMCs, thereby impacting the biological properties of vascular contraction, relaxation, and growth. Increasing evidence suggests that abnormal phenotypic changes in VSMCs contribute to the initiation of vascular diseases, including atherosclerosis. Therefore, understanding the factors governing phenotypic alterations in VSMCs and elucidating the underlying mechanisms can provide crucial insights for refining interventions targeted at vascular diseases. Additionally, the varying levels of different types of sex hormones in the human body, influenced by sex and age, may also affect the phenotypic conversion of VSMCs. This review aims to explore the influence of sex hormones on the phenotypic switching of VSMCs and the development of associated vascular diseases in the human body.
Collapse
Affiliation(s)
- Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xin Luo
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
19
|
Zisser L, Binder CJ. Extracellular Vesicles as Mediators in Atherosclerotic Cardiovascular Disease. J Lipid Atheroscler 2024; 13:232-261. [PMID: 39355407 PMCID: PMC11439751 DOI: 10.12997/jla.2024.13.3.232] [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: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/03/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial intima, characterized by accumulation of lipoproteins and accompanying inflammation, leading to the formation of plaques that eventually trigger occlusive thrombotic events, such as myocardial infarction and ischemic stroke. Although many aspects of plaque development have been elucidated, the role of extracellular vesicles (EVs), which are lipid bilayer-delimited vesicles released by cells as mediators of intercellular communication, has only recently come into focus of atherosclerosis research. EVs comprise several subtypes that may be differentiated by their size, mode of biogenesis, or surface marker expression and cargo. The functional effects of EVs in atherosclerosis depend on their cellular origin and the specific pathophysiological context. EVs have been suggested to play a role in all stages of plaque formation. In this review, we highlight the known mechanisms by which EVs modulate atherogenesis and outline current limitations and challenges in the field.
Collapse
Affiliation(s)
- Lucia Zisser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
20
|
Karabayas M, Ibrahim HE, Roelofs AJ, Reynolds G, Kidder D, De Bari C. Vascular disease persistence in giant cell arteritis: are stromal cells neglected? Ann Rheum Dis 2024; 83:1100-1109. [PMID: 38684323 PMCID: PMC11420755 DOI: 10.1136/ard-2023-225270] [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: 11/11/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Giant cell arteritis (GCA), the most common systemic vasculitis, is characterised by aberrant interactions between infiltrating and resident cells of the vessel wall. Ageing and breach of tolerance are prerequisites for GCA development, resulting in dendritic and T-cell dysfunction. Inflammatory cytokines polarise T-cells, activate resident macrophages and synergistically enhance vascular inflammation, providing a loop of autoreactivity. These events originate in the adventitia, commonly regarded as the biological epicentre of the vessel wall, with additional recruitment of cells that infiltrate and migrate towards the intima. Thus, GCA-vessels exhibit infiltrates across the vascular layers, with various cytokines and growth factors amplifying the pathogenic process. These events activate ineffective repair mechanisms, where dysfunctional vascular smooth muscle cells and fibroblasts phenotypically shift along their lineage and colonise the intima. While high-dose glucocorticoids broadly suppress these inflammatory events, they cause well known deleterious effects. Despite the emerging targeted therapeutics, disease relapse remains common, affecting >50% of patients. This may reflect a discrepancy between systemic and local mediators of inflammation. Indeed, temporal arteries and aortas of GCA-patients can show immune-mediated abnormalities, despite the treatment induced clinical remission. The mechanisms of persistence of vascular disease in GCA remain elusive. Studies in other chronic inflammatory diseases point to the fibroblasts (and their lineage cells including myofibroblasts) as possible orchestrators or even effectors of disease chronicity through interactions with immune cells. Here, we critically review the contribution of immune and stromal cells to GCA pathogenesis and analyse the molecular mechanisms by which these would underpin the persistence of vascular disease.
Collapse
Affiliation(s)
- Maira Karabayas
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Hafeez E Ibrahim
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Anke J Roelofs
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Gary Reynolds
- Centre for Immunology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Dana Kidder
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Cosimo De Bari
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
21
|
Wang B, Cui K, Zhu B, Dong Y, Wang D, Singh B, Wu H, Li K, Eisa-Beygi S, Sun Y, Wong S, Cowan DB, Chen Y, Du M, Chen H. Epsins oversee smooth muscle cell reprograming by influencing master regulators KLF4 and OCT4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574714. [PMID: 39131381 PMCID: PMC11312448 DOI: 10.1101/2024.01.08.574714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Smooth muscle cells in major arteries play a crucial role in regulating coronary artery disease. Conversion of smooth muscle cells into other adverse cell types in the artery propels the pathogenesis of the disease. Curtailing artery plaque buildup by modulating smooth muscle cell reprograming presents us a new opportunity to thwart coronary artery disease. Here, our report how Epsins, a family of endocytic adaptor proteins oversee the smooth muscle cell reprograming by influencing master regulators OCT4 and KLF4. Using single-cell RNA sequencing, we characterized the phenotype of modulated smooth muscle cells in mouse atherosclerotic plaque and found that smooth muscle cells lacking epsins undergo profound reprogramming into not only beneficial myofibroblasts but also endothelial cells for injury repair of diseased endothelium. Our work lays concrete groundwork to explore an uncharted territory as we show that depleting Epsins bolsters smooth muscle cells reprograming to endothelial cells by augmenting OCT4 activity but restrain them from reprograming to harmful foam cells by destabilizing KLF4, a master regulator of adverse reprograming of smooth muscle cells. Moreover, the expression of Epsins in smooth muscle cells positively correlates with the severity of both human and mouse coronary artery disease. Integrating our scRNA-seq data with human Genome-Wide Association Studies (GWAS) identifies pivotal roles Epsins play in smooth muscle cells in the pathological process leading to coronary artery disease. Our findings reveal a previously unexplored direction for smooth muscle cell phenotypic modulation in the development and progression of coronary artery disease and unveil Epsins and their downstream new targets as promising novel therapeutic targets for mitigating metabolic disorders.
Collapse
Affiliation(s)
- Beibei Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Kui Cui
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Bo Zhu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yunzhou Dong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Donghai Wang
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Bandana Singh
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Hao Wu
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yong Sun
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yabing Chen
- Department of Pathology, Birmingham, AL 35294, USA; University of Alabama at Birmingham, and the Birmingham Veterans Affairs Medical Center, Birmingham, AL 35294, USA
| | - Mulong Du
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital and Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
22
|
Han X, Li W, Chen C, Liu J, Sun J, Wang F, Wang C, Mu J, Gu X, Liu F, Xie H, Yang S, Shen C. Genetic variants and mRNA expression of KLF4 and KLF5 with hypertension: A combination of case-control study and cohort study. J Biomed Res 2024; 38:1-10. [PMID: 39187911 DOI: 10.7555/jbr.38.20240208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024] Open
Abstract
Hypertension (HT) is a major risk factor for cardiovascular diseases. Krüppel-like factors (KLFs) are important transcription factors in eukaryotes. Studies have reported that KLF4 and KLF5 are correlated with several cardiovascular diseases, whereas population studies for associations between HT and KLF4 or KLF5 have been rarely reported. Thus, the current study aimed to examines the association of genetic variants and mRNA expression levels of KLF4 and KLF5 with HT, as well as the effect of antihypertensive drugs on the expression levels. The associations of one single-nucleotide polymorphism (SNP) in KLF4 and three SNPs in KLF5 with HT were investigated using a combination of case-control and cohort studies. The study population were selected from a community-based population cohort in four different regions of Jiangsu Province. Risks of HT were estimated through logistic and Cox regression analyses, respectively. In addition, mRNA expression levels of KLF4 and KLF5 were measured in 246 controls and 385 HT cases selected from the cohort study as mentioned above. Among the HT cases, 263 were not taking antihypertensive drugs [AHD(-)] and 122 were taking antihypertensive drugs [AHD(+)]. In the case-control study, SNP rs9573096 (C>T) in KLF5 was significantly associated with an increased risk of HT in the additive model (adjusted odds ratio [OR], 1.106; 95% confidence interval [CI], 1.009 to 1.212). In the cohort study of the normotensive population, rs9573096 in KLF5 was also significantly associated with an increased risk of HT in the additive model (adjusted hazards ratio [HR], 1.199; 95% CI, 1.070 to 1.344). KLF4 and KLF5 mRNA expression levels were significantly higher in the AHD(-) group than in the control group ( P < 0.05), but lower in the AHD(+) group than in the AHD(-) group ( P < 0.05). The current study demonstrated the associations of KLF4 and KLF5 genetic variants with hypertension, and the indicative discriminations of mRNA expression levels of KLF4 and KLF5 for risk of hypertension and antihypertensive treatment.
Collapse
Affiliation(s)
- Xu Han
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wen Li
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Changying Chen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiahui Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Junxiang Sun
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, Jiangsu 214200, China
| | - Feifan Wang
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chao Wang
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jialing Mu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xincheng Gu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Fangyuan Liu
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Hankun Xie
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Song Yang
- Department of Cardiology, Affiliated Yixing People's Hospital of Jiangsu University, People's Hospital of Yixing City, Yixing, Jiangsu 214200, China
| | - Chong Shen
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
23
|
Quelquejay H, Al-Rifai R, Silvestro M, Vandestienne M, Ferreira I, Mirault T, Henrion D, Zhong X, Santos-Zas I, Goudot G, Alayrac P, Robidel E, Autret G, Balvay D, Taleb S, Tedgui A, Boulanger CM, Zernecke A, Saliba AE, Hadchouel J, Ramkhelawon B, Cochain C, Bergaya S, Jeunemaitre X, Ait-Oufella H. L-Wnk1 Deletion in Smooth Muscle Cells Causes Aortitis and Inflammatory Shift. Circ Res 2024; 135:488-502. [PMID: 38979610 DOI: 10.1161/circresaha.124.324366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND The long isoform of the Wnk1 (with-no-lysine [K] kinase 1) is a ubiquitous serine/threonine kinase, but its role in vascular smooth muscle cells (VSMCs) pathophysiology remains unknown. METHODS AngII (angiotensin II) was infused in Apoe-/- to induce experimental aortic aneurysm. Mice carrying an Sm22-Cre allele were cross-bred with mice carrying a floxed Wnk1 allele to specifically investigate the functional role of Wnk1 in VSMCs. RESULTS Single-cell RNA-sequencing of the aneurysmal abdominal aorta from AngII-infused Apoe-/- mice revealed that VSMCs that did not express Wnk1 showed lower expression of contractile phenotype markers and increased inflammatory activity. Interestingly, WNK1 gene expression in VSMCs was decreased in human abdominal aortic aneurysm. Wnk1-deficient VSMCs lost their contractile function and exhibited a proinflammatory phenotype, characterized by the production of matrix metalloproteases, as well as cytokines and chemokines, which contributed to local accumulation of inflammatory macrophages, Ly6Chi monocytes, and γδ T cells. Sm22Cre+Wnk1lox/lox mice spontaneously developed aortitis in the infrarenal abdominal aorta, which extended to the thoracic area over time without any negative effect on long-term survival. AngII infusion in Sm22Cre+Wnk1lox/lox mice aggravated the aortic disease, with the formation of lethal abdominal aortic aneurysms. Pharmacological blockade of γδ T-cell recruitment using neutralizing anti-CXCL9 (anti-CXC motif chemokine ligand 9) antibody treatment, or of monocyte/macrophage using Ki20227, a selective inhibitor of CSF1 receptor, attenuated aortitis. Wnk1 deletion in VSMCs led to aortic wall remodeling with destruction of elastin layers, increased collagen content, and enhanced local TGF-β (transforming growth factor-beta) 1 expression. Finally, in vivo TGF-β blockade using neutralizing anti-TGF-β antibody promoted saccular aneurysm formation and aorta rupture in Sm22 Cre+ Wnk1lox/lox mice but not in control animals. CONCLUSION Wnk1 is a key regulator of VSMC function. Wnk1 deletion promotes VSMC phenotype switch toward a pathogenic proinflammatory phenotype, orchestrating deleterious vascular remodeling and spontaneous severe aortitis in mice.
Collapse
MESH Headings
- Animals
- Aortitis/genetics
- Aortitis/metabolism
- Aortitis/pathology
- Mice
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Angiotensin II
- Aortic Aneurysm, Abdominal/genetics
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Humans
- WNK Lysine-Deficient Protein Kinase 1/genetics
- WNK Lysine-Deficient Protein Kinase 1/metabolism
- Mice, Inbred C57BL
- Male
- Cells, Cultured
- Mice, Knockout, ApoE
- Disease Models, Animal
- Inflammation/metabolism
- Inflammation/genetics
- Inflammation/pathology
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
Collapse
Affiliation(s)
- Helene Quelquejay
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Rida Al-Rifai
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Michele Silvestro
- Division of Vascular and Endovascular Surgery, Department of Surgery and Department of Cell Biology, New York University Langone Medical Center (M.S., B.R.)
| | - Marie Vandestienne
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Irmine Ferreira
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Tristan Mirault
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Daniel Henrion
- MITOVASC Department, Team 2 (CarMe), ICAT SFR (Interactions Cellulaires et Applications Thérapeutiques Structure Fédérale de Recherche), University of Angers, Inserm U1083, France (D.H.)
| | - Xiaodan Zhong
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Icia Santos-Zas
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
- Laboratorio de Endocrinología Celular, Área de Endocrinología Molecular y Celular Instituto de Investigación Sanitaria de Santiago, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain (I.S.-Z.)
| | - Guillaume Goudot
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Paul Alayrac
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Estelle Robidel
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Gwennhael Autret
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Daniel Balvay
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Soraya Taleb
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Alain Tedgui
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Chantal M Boulanger
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (A.Z., C.C.)
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany (A.-E.S.)
| | - Juliette Hadchouel
- Inserm UMRS 1155, Tenon Hospital (J.H.), Sorbonne Université, Paris, France
| | - Bhama Ramkhelawon
- Division of Vascular and Endovascular Surgery, Department of Surgery and Department of Cell Biology, New York University Langone Medical Center (M.S., B.R.)
| | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Germany (A.Z., C.C.)
| | - Sonia Bergaya
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Xavier Jeunemaitre
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
| | - Hafid Ait-Oufella
- Université de Paris, Inserm U970, Paris-Cardiovascular Research Center, France (H.Q., R.A.-R., M.V., I.F., T.M., X.Z., I.S.-Z., G.G., P.A., E.R., G.A., D.B., S.T., A.T., C.M.B., S.B., X.J., H.A.-O.)
- Medical Intensive Care Unit, Hôpital Saint-Antoine, AP-HP (Assistance Publique- Hôpitaux de Paris) (H.A.-O.), Sorbonne Université, Paris, France
| |
Collapse
|
24
|
Liang G, Lv XF, Huang W, Jin YJ, Roquid KA, Kawase H, Offermanns S. Loss of Smooth Muscle Tenascin-X Inhibits Vascular Remodeling Through Increased TGF-β Signaling. Arterioscler Thromb Vasc Biol 2024; 44:1748-1763. [PMID: 38934115 DOI: 10.1161/atvbaha.123.321067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Vascular smooth muscle cells (VSMCs) are highly plastic. Vessel injury induces a phenotypic transformation from differentiated to dedifferentiated VSMCs, which involves reduced expression of contractile proteins and increased production of extracellular matrix and inflammatory cytokines. This transition plays an important role in several cardiovascular diseases such as atherosclerosis, hypertension, and aortic aneurysm. TGF-β (transforming growth factor-β) is critical for VSMC differentiation and to counterbalance the effect of dedifferentiating factors. However, the mechanisms controlling TGF-β activity and VSMC phenotypic regulation under in vivo conditions are poorly understood. The extracellular matrix protein TN-X (tenascin-X) has recently been shown to bind TGF-β and to prevent it from activating its receptor. METHODS We studied the role of TN-X in VSMCs in various murine disease models using tamoxifen-inducible SMC-specific knockout and adeno-associated virus-mediated knockdown. RESULTS In hypertensive and high-fat diet-fed mice, after carotid artery ligation as well as in human aneurysmal aortae, expression of Tnxb, the gene encoding TN-X, was increased in VSMCs. Mice with smooth muscle cell-specific loss of TN-X (SMC-Tnxb-KO) showed increased TGF-β signaling in VSMCs, as well as upregulated expression of VSMC differentiation marker genes during vascular remodeling compared with controls. SMC-specific TN-X deficiency decreased neointima formation after carotid artery ligation and reduced vessel wall thickening during Ang II (angiotensin II)-induced hypertension. SMC-Tnxb-KO mice lacking ApoE showed reduced atherosclerosis and Ang II-induced aneurysm formation under high-fat diet. Adeno-associated virus-mediated SMC-specific expression of short hairpin RNA against Tnxb showed similar beneficial effects. Treatment with an anti-TGF-β antibody or additional SMC-specific loss of the TGF-β receptor reverted the effects of SMC-specific TN-X deficiency. CONCLUSIONS In summary, TN-X critically regulates VSMC plasticity during vascular injury by inhibiting TGF-β signaling. Our data indicate that inhibition of vascular smooth muscle TN-X may represent a strategy to prevent and treat pathological vascular remodeling.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/prevention & control
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/metabolism
- Carotid Artery Injuries/genetics
- Cells, Cultured
- Diet, High-Fat
- Disease Models, Animal
- Hypertension/metabolism
- Hypertension/pathology
- Hypertension/physiopathology
- Hypertension/genetics
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenotype
- Signal Transduction
- Tenascin/metabolism
- Tenascin/genetics
- Tenascin/deficiency
- Transforming Growth Factor beta/metabolism
- Vascular Remodeling
Collapse
Affiliation(s)
- Guozheng Liang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Xiao-Fei Lv
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China (X.-F.L.)
| | - Wei Huang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Young-June Jin
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Kenneth Anthony Roquid
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Haruya Kawase
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany (G.L., X.-F.L., W.H., Y.-J.J., K.A.R., H.K., S.O.)
- Center for Molecular Medicine, Goethe University Frankfurt, Germany (S.O.)
- Cardiopulmonary Institute, Bad Nauheim, Germany (S.O.)
- German Center for Cardiovascular Research, Bad Nauheim, Germany (S.O.)
| |
Collapse
|
25
|
Vartak T, Giardini E, Kelly D, Moran B, Kennedy C, Barry M, Godson C, Brennan E. Induction of let-7d-5p miRNA modulates aortic smooth muscle inflammatory signaling and phenotypic switching. Atherosclerosis 2024; 395:117573. [PMID: 38796407 DOI: 10.1016/j.atherosclerosis.2024.117573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND AND AIMS Activation of vascular smooth muscle cell inflammation is recognised as an important early driver of vascular disease. We have previously identified the let-7 miRNA family as important regulators of inflammation in in vitro and in vivo models of atherosclerosis. Here we investigated a dual statin/let-7d-5p miRNA combination therapy approach to target human aortic SMC (HAoSMC) activation and inflammation. METHODS In vitro studies using primary HAoSMCs were performed to investigate the effects of let-7d-5p miRNA overexpression and inhibition. HAoSMCs were treated with combinations of the inflammatory cytokine tumor necrosis factor-α (TNF-α), and atorvastatin or lovastatin. HAoSMC Bulk RNA-seq transcriptomics of HAoSMCs revealed downstream regulatory networks modulated by let-7d-5p miRNA overexpression and statins. Proteome profiler cytokine array, Western blotting and quantitative PCR analyses were performed on HAoSMCs to validate key findings. RESULTS Let-7d-5p overexpression significantly attenuated TNF-α-induced upregulation of IL-6, ICAM1, VCAM1, CCL2, CD68, MYOCD gene expression in HAoSMCs (p<0.05). Statins (atorvastatin, lovastatin) significantly attenuated inflammatory gene expression and upregulated Let-7d levels in HAoSMCs (p<0.05). Bulk RNA-seq analysis of a dual Let-7d-5p overexpression/statin therapy in HAoSMCs revealed that let-7d-5p activation and statins converge on key inflammatory pathways (IL-6, IL-1β, TNF-α, IFN-γ). Let-7d-5p overexpression led to reduced expression of the ox-LDL receptor OLR1, and this was associated with lower ox-LDL uptake in HAoSMCs. In silico analysis of smooth muscle cell phenotypic switching shows that overexpression of let-7d-5p in HAoSMCs maintains a contractile phenotype. CONCLUSIONS Targeting the Let-7 network alongside statins can modulate HAoSMC activation and attenuate key inflammatory pathway signals.
Collapse
Affiliation(s)
- Tanwi Vartak
- Diabetes Complications Research Centre, Conway Institute & UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Elena Giardini
- Diabetes Complications Research Centre, Conway Institute & UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Daniel Kelly
- Diabetes Complications Research Centre, Conway Institute & UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Bruce Moran
- St. Vincent's University Hospital, Dublin, Ireland
| | - Ciarán Kennedy
- Diabetes Complications Research Centre, Conway Institute & UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Mary Barry
- Department of Vascular Surgery, St. Vincent's University Hospital, Dublin, Ireland
| | - Catherine Godson
- Diabetes Complications Research Centre, Conway Institute & UCD School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Eoin Brennan
- Diabetes Complications Research Centre, Conway Institute & UCD School of Medicine, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
26
|
Yang S, Zhao Y, Cao S, Liu X, Feng M, Chen Y, Ma C, Zhan T, Zhang Q, Jia H, Zhao Y, Tong M, Yu Y, Liu X, Yang B, Zhang Y. Kanglexin counters vascular smooth muscle cell dedifferentiation and associated arteriosclerosis through inhibiting PDGFR. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155704. [PMID: 38759316 DOI: 10.1016/j.phymed.2024.155704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Dysregulation of vascular smooth muscle cell (VSMC) function leads to a variety of diseases such as atherosclerosis and hyperplasia after injury. However, antiproliferative drug targeting VSMC exhibits poor specificity. Therefore, there is an urgent to develop highly specific antiproliferative drugs to prevention and treatment VSMC dedifferentiation associated arteriosclerosis. Kanglexin (KLX), a new anthraquinone compound designed by our team, has potential to regulate VSMC phenotype according to the physicochemical properties. PURPOSE This project aims to evaluate the therapeutic role of KLX in VSMC dedifferentiation and atherosclerosis, neointimal formation and illustrates the underlying molecular mechanism. METHODS In vivo, the ApoE-/- mice were fed with high-fat diet (HFD) for a duration of 13 weeks to establish the atherosclerotic model. And rat carotid artery injury model was performed to establish the neointimal formation model. In vitro, PDGF-BB was used to induce VSMC dedifferentiation. RESULTS We found that KLX ameliorated the atherosclerotic progression including atherosclerotic lesion formation, lipid deposition and collagen deposition in aorta and aortic sinus in atherosclerotic mouse model. In addition, The administration of KLX effectively ameliorated neointimal formation in the carotid artery following balloon injury in SD rats. The findings derived from molecular docking and surface plasmon resonance (SPR) experiments unequivocally demonstrate that KLX had potential to bind PDGFR-β. Mechanism research work proved that KLX prevented VSMC proliferation, migration and dedifferentiation via activating the PDGFR-β-MEK -ERK-ELK-1/KLF4 signaling pathway. CONCLUSION Collectively, we demonstrated that KLX effectively attenuated the progression of atherosclerosis in ApoE-/- mice and carotid arterial neointimal formation in SD rats by inhibiting VSMC phenotypic conversion via PDGFR-β-MEK-ERK-ELK-1/KLF4 signaling. KLX exhibits promising potential as a viable therapeutic agent for the treatment of VSMC phenotype conversion associated arteriosclerosis.
Collapse
Affiliation(s)
- Shuang Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yixiu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Shifeng Cao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xinxin Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Min Feng
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yi Chen
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Chunyue Ma
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Tingting Zhan
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Qi Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Honglin Jia
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yu Zhao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Ming Tong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuanyuan Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Xue Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Baofeng Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China.
| | - Yan Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
27
|
Li J, Lan T, Guo Q, Zhang C, Lu X, Hu X, Shen X, Zhang Y. Mitochondria-Targeted Natural Antioxidant Nanosystem for Diabetic Vascular Calcification Therapy. Biomacromolecules 2024; 25:4329-4343. [PMID: 38833553 DOI: 10.1021/acs.biomac.4c00375] [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: 06/06/2024]
Abstract
The development of nanotherapy targeting mitochondria to alleviate oxidative stress is a critical therapeutic strategy for vascular calcification (VC) in diabetes. In this study, we engineered mitochondria-targeted nanodrugs (T4O@TPP/PEG-PLGA) utilizing terpinen-4-ol (T4O) as a natural antioxidant and mitochondrial protector, PEG-PLGA as the nanocarrier, and triphenylphosphine (TPP) as the mitochondrial targeting ligand. In vitro assessments demonstrated enhanced cellular uptake of T4O@TPP/PEG-PLGA, with effective mitochondrial targeting. This nanodrug successfully reduced oxidative stress induced by high glucose levels in vascular smooth muscle cells. In vivo studies showed prolonged retention of the nanomaterials in the thoracic aorta for up to 24 h. Importantly, experiments in diabetic VC models underscored the potent antioxidant properties of T4O@TPP/PEG-PLGA, as evidenced by its ability to mitigate VC and restore mitochondrial morphology. These results suggest that these nanodrugs could be a promising strategy for managing diabetic VC.
Collapse
Affiliation(s)
- Jinjin Li
- The Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Tianyu Lan
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- College of Ethnic Medicine, Guizhou Minzu University, Guiyang 550025, Guizhou, China
| | - Qianqian Guo
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Chuang Zhang
- The Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Xin Lu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Xiaoxia Hu
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| | - Yanyan Zhang
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Department of Pharmacology of Materia Medica (The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, University Town, Guian New District, Guiyang 550025, Guizhou, China
| |
Collapse
|
28
|
Kwartler CS, Pinelo JEE. Use of iPSC-Derived Smooth Muscle Cells to Model Physiology and Pathology. Arterioscler Thromb Vasc Biol 2024; 44:1523-1536. [PMID: 38695171 PMCID: PMC11209779 DOI: 10.1161/atvbaha.123.319703] [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: 06/28/2024]
Abstract
The implementation of human induced pluripotent stem cell (hiPSC) models has introduced an additional tool for identifying molecular mechanisms of disease that complement animal models. Patient-derived or CRISPR/Cas9-edited induced pluripotent stem cells differentiated into smooth muscle cells (SMCs) have been leveraged to discover novel mechanisms, screen potential therapeutic strategies, and model in vivo development. The field has evolved over almost 15 years of research using hiPSC-SMCs and has made significant strides toward overcoming initial challenges such as the lineage specificity of SMC phenotypes. However, challenges both specific (eg, the lack of specific markers to thoroughly validate hiPSC-SMCs) and general (eg, a lack of transparency and consensus around methodology in the field) remain. In this review, we highlight the recent successes and remaining challenges of the hiPSC-SMC model.
Collapse
Affiliation(s)
- Callie S. Kwartler
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Jose Emiliano Esparza Pinelo
- Division of Medical Genetics, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
29
|
Towler DA. Parathyroid hormone-PTH1R signaling in cardiovascular disease and homeostasis. Trends Endocrinol Metab 2024; 35:648-660. [PMID: 38429163 PMCID: PMC11233248 DOI: 10.1016/j.tem.2024.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Primary hyperparathyroidism (pHPT) afflicts our aging population with an incidence approaching 50 per 100 000 patient-years at a female:male ratio of ~3:1. Decisions surrounding surgical management are currently driven by age, hypercalcemia severity, presence of osteoporosis, renal insufficiency, or hypercalciuria with or without nephrolithiasis. Cardiovascular (CV) disease (CVD) is not systematically considered. This is notable since the parathyroid hormone (PTH) 1 receptor (PTH1R) is biologically active in the vasculature, and adjusted CV mortality risk is increased almost threefold in individuals with pHPT who do not meet contemporary recommendations for surgical cure. We provide an overview of epidemiology, pharmacology, and physiology that highlights the need to: (i) identify biomarkers that establish a healthy 'set point' for CV PTH1R signaling tone; (ii) better understand the pharmacokinetic-pharmacodynamic (PK-PD) relationships of PTH1R ligands in CV homeostasis; and (iii) incorporate CVD risk assessment into the management of hyperparathyroidism.
Collapse
Affiliation(s)
- Dwight A Towler
- Department of Internal Medicine - Endocrine Division, Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
30
|
Berkholz J, Karle W. Unravelling the molecular interplay: SUMOylation, PML nuclear bodies and vascular cell activity in health and disease. Cell Signal 2024; 119:111156. [PMID: 38574938 DOI: 10.1016/j.cellsig.2024.111156] [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: 01/01/2024] [Revised: 03/23/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
In the seemingly well-researched field of vascular research, there are still many underestimated factors and molecular mechanisms. In recent years, SUMOylation has become increasingly important. SUMOylation is a post-translational modification in which small ubiquitin-related modifiers (SUMO) are covalently attached to target proteins. Sites where these SUMO modification processes take place in the cell nucleus are PML nuclear bodies (PML-NBs) - multiprotein complexes with their essential main component and organizer, the PML protein. PML and SUMO, either alone or as partners, influence a variety of cellular processes, including regulation of transcription, senescence, DNA damage response and defence against microorganisms, and are involved in innate immunity and inflammatory responses. They also play an important role in maintaining homeostasis in the vascular system and in pathological processes leading to the development and progression of cardiovascular diseases. This review summarizes information about the function of SUMO(ylation) and PML(-NBs) in the human vasculature from angiogenesis to disease and highlights their clinical potential as drug targets.
Collapse
Affiliation(s)
- Janine Berkholz
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany; DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany.
| | - Weronika Karle
- Institute of Physiology, Charité - Universitätsmedizin, Berlin, Germany
| |
Collapse
|
31
|
Martinez AN, Tortelote GG, Pascale CL, Ekanem UOI, Leite APDO, McCormack IG, Dumont AS. Dimethyl Fumarate Mediates Sustained Vascular Smooth Muscle Cell Remodeling in a Mouse Model of Cerebral Aneurysm. Antioxidants (Basel) 2024; 13:773. [PMID: 39061841 PMCID: PMC11274241 DOI: 10.3390/antiox13070773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/28/2024] Open
Abstract
Cerebral aneurysms (CA) are a type of vascular disease that causes significant morbidity and mortality with rupture. Dysfunction of the vascular smooth muscle cells (VSMCs) from circle of Willis (CoW) vessels mediates CA formation, as they are the major cell type of the arterial wall and play a role in maintaining vessel integrity. Dimethyl fumarate (DMF), a first-line oral treatment for relapsing-remitting multiple sclerosis, has been shown to inhibit VSMC proliferation and reduce CA formation in a mouse model. Potential unwanted side effects of DMF on VSMC function have not been investigated yet. The present study characterizes the impact of DMF on VSMC using single-cell RNA-sequencing (scRNA-seq) in CoW vessels following CA induction and further explores its role in mitochondrial function using in vitro VSMC cultures. Two weeks of DMF treatment following CA induction impaired the transcription of the glutathione redox system and downregulated mitochondrial respiration genes in VSMCs. In vitro, DMF treatment increased lactate formation and enhanced the mitochondrial production of reactive oxygen species (ROS). These effects rendered VSMCs vulnerable to oxidative stress and led to mitochondrial dysfunction and enhancement of apoptosis. Taken together, our data support the concept that the DMF-mediated antiproliferative effect on VSMCs is linked to disturbed antioxidative functions resulting in altered mitochondrial metabolism. This negative impact of DMF treatment on VSMCs may be linked to preexisting alterations of cerebrovascular function due to renal hypertension. Therefore, before severe adverse effects emerge, it would be clinically relevant to develop indices or biomarkers linked to this disturbed antioxidative function to monitor patients undergoing DMF treatment.
Collapse
Affiliation(s)
- Alejandra N. Martinez
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Giovane G. Tortelote
- Department of Pediatrics, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| | - Crissey L. Pascale
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Uduak-Obong I. Ekanem
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Ana Paula de O. Leite
- Department of Pharmacology, The Tulane Center for Sex-Based Biology and Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Isabella G. McCormack
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| | - Aaron S. Dumont
- Department of Neurosurgery, The Tulane Center for Clinical Neurosciences, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA 70012, USA (A.S.D.)
| |
Collapse
|
32
|
Lu G, Guo H, Zhang Y, Zhang M, Zhang T, Hu G, Zhang Q. Graphene Far-Infrared Irradiation Can Effectively Relieve the Blood Pressure Level of Rat Untr-HT in Primary Hypertension. Int J Mol Sci 2024; 25:6675. [PMID: 38928382 PMCID: PMC11204347 DOI: 10.3390/ijms25126675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Graphene, when electrified, generates far-infrared radiation within the wavelength range of 4 μm to 14 μm. This range closely aligns with the far-infrared band (3 μm to 15 μm), which produces unique physiological effects. Contraction and relaxation of vascular smooth muscle play a significant role in primary hypertension, involving the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate pathway and the renin-angiotensin-aldosterone system. This study utilized spontaneously hypertensive rats (SHRs) as an untr-HT to investigate the impact of far-infrared radiation at specific wavelengths generated by electrified graphene on vascular smooth muscle and blood pressure. After 7 weeks, the blood pressure of the untr-HT group rats decreased significantly with a notable reduction in the number of vascular wall cells and the thickness of the vascular wall, as well as a decreased ratio of vessel wall thickness to lumen diameter. Additionally, blood flow perfusion significantly increased, and the expression of F-actin in vascular smooth muscle myosin decreased significantly. Serum levels of angiotensin II (Ang-II) and endothelin 1 (ET-1) were significantly reduced, while nitric oxide synthase (eNOS) expression increased significantly. At the protein level, eNOS expression decreased significantly, while α-SMA expression increased significantly in aortic tissue. At the gene level, expressions of eNOS and α-SMA in aortic tissue significantly increased. Furthermore, the content of nitric oxide (NO) in the SHR's aortic tissue increased significantly. These findings confirm that graphene far-infrared radiation enhances microcirculation, regulates cytokines affecting vascular smooth muscle contraction, and modifies vascular morphology and smooth muscle phenotype, offering relief for primary hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qian Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China; (G.L.); (H.G.); (Y.Z.); (M.Z.); (T.Z.); (G.H.)
| |
Collapse
|
33
|
Li CX, Yue L. The Multifaceted Nature of Macrophages in Cardiovascular Disease. Biomedicines 2024; 12:1317. [PMID: 38927523 PMCID: PMC11201197 DOI: 10.3390/biomedicines12061317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/01/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
As the leading cause of mortality worldwide, cardiovascular disease (CVD) represents a variety of heart diseases and vascular disorders, including atherosclerosis, aneurysm, ischemic injury in the heart and brain, arrythmias, and heart failure. Macrophages, a diverse population of immune cells that can promote or suppress inflammation, have been increasingly recognized as a key regulator in various processes in both healthy and disease states. In healthy conditions, these cells promote the proper clearance of cellular debris, dead and dying cells, and provide a strong innate immune barrier to foreign pathogens. However, macrophages can play a detrimental role in the progression of disease as well, particularly those inflammatory in nature. This review will focus on the current knowledge regarding the role of macrophages in cardiovascular diseases.
Collapse
Affiliation(s)
- Cindy X. Li
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| | - Lixia Yue
- Department of Cell Biology, Pat and Jim Calhoun Cardiovascular Center, University of Connecticut Health Center, Farmington, CT 06030, USA;
- Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
34
|
Kang K, Sun C, Li H, Liu X, Deng J, Chen S, Zeng L, Chen J, Liu X, Kuang J, Xiang J, Cheng J, Liao X, Lin M, Zhang X, Zhan C, Liu S, Wang J, Niu Y, Liu C, Liang C, Zhu J, Liang S, Tang H, Gou D. N6-methyladenosine-driven miR-143/145-KLF4 circuit orchestrates the phenotypic switch of pulmonary artery smooth muscle cells. Cell Mol Life Sci 2024; 81:256. [PMID: 38866991 PMCID: PMC11335293 DOI: 10.1007/s00018-024-05304-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024]
Abstract
Pulmonary hypertension (PH) is characterized by vascular remodeling predominantly driven by a phenotypic switching in pulmonary artery smooth muscle cells (PASMCs). However, the underlying mechanisms for this phenotypic alteration remain incompletely understood. Here, we identified that RNA methyltransferase METTL3 is significantly elevated in the lungs of hypoxic PH (HPH) mice and rats, as well as in the pulmonary arteries (PAs) of HPH rats. Targeted deletion of Mettl3 in smooth muscle cells exacerbated hemodynamic consequences of hypoxia-induced PH and accelerated pulmonary vascular remodeling in vivo. Additionally, the absence of METTL3 markedly induced phenotypic switching in PASMCs in vitro. Mechanistically, METTL3 depletion attenuated m6A modification and hindered the processing of pri-miR-143/145, leading to a downregulation of miR-143-3p and miR-145-5p. Inhibition of hnRNPA2B1, an m6A mediator involved in miRNA maturation, similarly resulted in a significant reduction of miR-143-3p and miR-145-5p. We demonstrated that miR-145-5p targets Krüppel-like factor 4 (KLF4) and miR-143-3p targets fascin actin-bundling protein 1 (FSCN1) in PASMCs. The decrease of miR-145-5p subsequently induced an upregulation of KLF4, which in turn suppressed miR-143/145 transcription, establishing a positive feedback circuit between KLF4 and miR-143/145. This regulatory circuit facilitates the persistent suppression of contractile marker genes, thereby sustaining PASMC phenotypic switch. Collectively, hypoxia-induced upregulation of METTL3, along with m6A mediated regulation of miR-143/145, might serve as a protective mechanism against phenotypic switch of PASMCs. Our results highlight a potential therapeutic strategy targeting m6A modified miR-143/145-KLF4 loop in the treatment of PH.
Collapse
Affiliation(s)
- Kang Kang
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Chuannan Sun
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Hui Li
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Xiaojia Liu
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Jingyuan Deng
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Silei Chen
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Le Zeng
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Jiahao Chen
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Xinyi Liu
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Jiahao Kuang
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Jingjing Xiang
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Jingqian Cheng
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Xiaoyun Liao
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Mujin Lin
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Xingshi Zhang
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Chuzhi Zhan
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Sisi Liu
- Department of Biochemistry and Molecular Biology, Shenzhen University Medical School, Shenzhen, 518060, Guangdong, China
| | - Jun Wang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Yanqin Niu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Cuilian Liu
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Cai Liang
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Jinsheng Zhu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Shuxin Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Haiyang Tang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
| | - Deming Gou
- Shenzhen Key Laboratory of Microbial Genetic Engineering, Vascular Disease Research Center, College of Life Sciences and Oceanography, Guangdong Provincial Key Laboratory of Regional Immunity and Disease, Carson International Cancer Center, School of Medicine, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
35
|
Bramel EE, Camejo WAE, Creamer TJ, Restrepo L, Saqib M, Bagirzadeh R, Zeng A, Mitchell JT, Stein-O’Brien GL, Pedroza AJ, Fischbein MP, Dietz HC, MacFarlane EG. Intrinsic Gata4 expression sensitizes the aortic root to dilation in a Loeys-Dietz syndrome mouse model. RESEARCH SQUARE 2024:rs.3.rs-4420617. [PMID: 38883722 PMCID: PMC11177966 DOI: 10.21203/rs.3.rs-4420617/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Loeys-Dietz syndrome (LDS) is an aneurysm disorder caused by mutations that decrease transforming growth factor-β (TGF-β) signaling. Although aneurysms develop throughout the arterial tree, the aortic root is a site of heightened risk. To identify molecular determinants of this vulnerability, we investigated the heterogeneity of vascular smooth muscle cells (VSMCs) in the aorta of Tgfbr1 M318R/+ LDS mice by single cell and spatial transcriptomics. Reduced expression of components of the extracellular matrix-receptor apparatus and upregulation of stress and inflammatory pathways were observed in all LDS VSMCs. However, regardless of genotype, a subset of Gata4-expressing VSMCs predominantly located in the aortic root intrinsically displayed a less differentiated, proinflammatory profile. A similar population was also identified among aortic VSMCs in a human scRNAseq dataset. Postnatal VSMC-specific Gata4 deletion reduced aortic root dilation in LDS mice, suggesting that this factor sensitizes the aortic root to the effects of impaired TGF-β signaling.
Collapse
Affiliation(s)
- Emily E. Bramel
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wendy A. Espinoza Camejo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Tyler J. Creamer
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leda Restrepo
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzna Saqib
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rustam Bagirzadeh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anthony Zeng
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jacob T. Mitchell
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Predoctoral Training in Human Genetics and Genomics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Genevieve L. Stein-O’Brien
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Albert J. Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Harry C. Dietz
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elena Gallo MacFarlane
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
36
|
Zhang R, Wang H, Cheng X, Fan K, Gao T, Qi X, Gao S, Zheng G, Dong H. High estrogen induces trans-differentiation of vascular smooth muscle cells to a macrophage-like phenotype resulting in aortic inflammation via inhibiting VHL/HIF1a/KLF4 axis. Aging (Albany NY) 2024; 16:9876-9898. [PMID: 38843385 PMCID: PMC11210252 DOI: 10.18632/aging.205904] [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: 12/15/2023] [Accepted: 04/22/2024] [Indexed: 06/22/2024]
Abstract
Estrogen is thought to have a role in slowing down aging and protecting cardiovascular and cognitive function. However, high doses of estrogen are still positively associated with autoimmune diseases and tumors with systemic inflammation. First, we administered exogenous estrogen to female mice for three consecutive months and found that the aorta of mice on estrogen develops inflammatory manifestations similar to Takayasu arteritis (TAK). Then, in vitro estrogen intervention was performed on mouse aortic vascular smooth muscle cells (MOVAS cells). Stimulated by high concentrations of estradiol, MOVAS cells showed decreased expression of contractile phenotypic markers and increased expression of macrophage-like phenotypic markers. This shift was blocked by tamoxifen and Krüppel-like factor 4 (KLF4) inhibitors and enhanced by Von Hippel-Lindau (VHL)/hypoxia-inducible factor-1α (HIF-1α) interaction inhibitors. It suggests that estrogen-targeted regulation of the VHL/HIF-1α/KLF4 axis induces phenotypic transformation of vascular smooth muscle cells (VSMC). In addition, estrogen-regulated phenotypic conversion of VSMC to macrophages is a key mechanism of estrogen-induced vascular inflammation, which justifies the risk of clinical use of estrogen replacement therapy.
Collapse
MESH Headings
- Kruppel-Like Factor 4
- Animals
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Macrophages/metabolism
- Macrophages/drug effects
- Mice
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Female
- Estrogens/pharmacology
- Von Hippel-Lindau Tumor Suppressor Protein/metabolism
- Von Hippel-Lindau Tumor Suppressor Protein/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Cell Transdifferentiation/drug effects
- Phenotype
- Aorta/pathology
- Aorta/drug effects
- Inflammation/metabolism
Collapse
Affiliation(s)
- Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xing Cheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Keyi Fan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Tingting Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaotong Qi
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Siqi Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoping Zheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
37
|
Karbasion N, Xu Y, Snider JC, Bersi MR. Primary Mouse Aortic Smooth Muscle Cells Exhibit Region- and Sex-Dependent Biological Responses In Vitro. J Biomech Eng 2024; 146:060904. [PMID: 38421345 PMCID: PMC11005860 DOI: 10.1115/1.4064965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/02/2024]
Abstract
Despite advancements in elucidating biological mechanisms of cardiovascular remodeling, cardiovascular disease (CVD) remains the leading cause of death worldwide. When stratified by sex, clear differences in CVD prevalence and mortality between males and females emerge. Regional differences in phenotype and biological response of cardiovascular cells are important for localizing the initiation and progression of CVD. Thus, to better understand region and sex differences in CVD presentation, we have focused on characterizing in vitro behaviors of primary vascular smooth muscle cells (VSMCs) from the thoracic and abdominal aorta of male and female mice. VSMC contractility was assessed by traction force microscopy (TFM; single cell) and collagen gel contraction (collective) with and without stimulation by transforming growth factor-beta 1 (TGF-β1) and cell proliferation was assessed by a colorimetric metabolic assay (MTT). Gene expression and TFM analysis revealed region- and sex-dependent behaviors, whereas collagen gel contraction was consistent across sex and aortic region under baseline conditions. Thoracic VSMCs showed a sex-dependent sensitivity to TGF-β1-induced collagen gel contraction (female > male; p = 0.025) and a sex-dependent proliferative response (female > male; p < 0.001) that was not apparent in abdominal VSMCs. Although primary VSMCs exhibit intrinsic region and sex differences in biological responses that may be relevant for CVD presentation, several factors-such as inflammation and sex hormones-were not included in this study. Such factors should be included in future studies of in vitro mechanobiological responses relevant to CVD differences in males and females.
Collapse
Affiliation(s)
- Niyousha Karbasion
- Department of Mechanical Engineering & Materials Science, Washington University at St. Louis, St. Louis, MO 63130
| | - Yujun Xu
- Department of Mechanical Engineering & Materials Science, Washington University at St. Louis, St. Louis, MO 63130
- Washington University in St. Louis
| | - J. Caleb Snider
- Department of Mechanical Engineering & Materials Science, Washington University at St. Louis, St. Louis, MO 63130
- Washington University in St. Louis
| | - Matthew R. Bersi
- Department of Mechanical Engineering & Materials Science, Washington University at St. Louis, St. Louis, MO 63130
| |
Collapse
|
38
|
Liu X, Zeng Q, Yang H, Li W, Chen Q, Yin K, Pan Z, Wang K, Luo M, Shu C, Zhou Z. Single-Nucleus Multiomic Analyses Identifies Gene Regulatory Dynamics of Phenotypic Modulation in Human Aneurysmal Aortic Root. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400444. [PMID: 38552156 PMCID: PMC11165511 DOI: 10.1002/advs.202400444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Indexed: 06/12/2024]
Abstract
Aortic root aneurysm is a potentially life-threatening condition that may lead to aortic rupture and is often associated with genetic syndromes, such as Marfan syndrome (MFS). Although studies with MFS animal models have provided valuable insights into the pathogenesis of aortic root aneurysms, this understanding of the transcriptomic and epigenomic landscape in human aortic root tissue remains incomplete. This knowledge gap has impeded the development of effective targeted therapies. Here, this study performs the first integrative analysis of single-nucleus multiomic (gene expression and chromatin accessibility) and spatial transcriptomic sequencing data of human aortic root tissue under healthy and MFS conditions. Cell-type-specific transcriptomic and cis-regulatory profiles in the human aortic root are identified. Regulatory and spatial dynamics during phenotypic modulation of vascular smooth muscle cells (VSMCs), the cardinal cell type, are delineated. Moreover, candidate key regulators driving the phenotypic modulation of VSMC, such as FOXN3, TEAD1, BACH2, and BACH1, are identified. In vitro experiments demonstrate that FOXN3 functions as a novel key regulator for maintaining the contractile phenotype of human aortic VSMCs through targeting ACTA2. These findings provide novel insights into the regulatory and spatial dynamics during phenotypic modulation in the aneurysmal aortic root of humans.
Collapse
Affiliation(s)
- Xuanyu Liu
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Qingyi Zeng
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Hang Yang
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Wenke Li
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Qianlong Chen
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Kunlun Yin
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Zihang Pan
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Kai Wang
- Department of Physiology and PathophysiologySchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingPeking UniversityBeijing100191China
| | - Mingyao Luo
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
- Center of Vascular SurgeryFuwai HospitalChinese Academy of Medical SciencesBeijing100037China
- Department of Vascular SurgeryFuwai Yunnan Cardiovascular HospitalAffiliated Cardiovascular Hospital of Kunming Medical UniversityKunmingYunnan650102China
- Department of Vascular SurgeryCentral‐China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
| | - Chang Shu
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
- Center of Vascular SurgeryFuwai HospitalChinese Academy of Medical SciencesBeijing100037China
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular DiseaseNational Center for Cardiovascular DiseasesBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesCenter of Laboratory MedicineFuwai HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| |
Collapse
|
39
|
Xu F, Chen H, Zhou C, Zang T, Wang R, Shen S, Li C, Yu Y, Pei Z, Shen L, Qian J, Ge J. Targeting deubiquitinase OTUB1 protects vascular smooth muscle cells in atherosclerosis by modulating PDGFRβ. Front Med 2024; 18:465-483. [PMID: 38644399 DOI: 10.1007/s11684-024-1056-8] [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: 07/20/2023] [Accepted: 12/04/2023] [Indexed: 04/23/2024]
Abstract
Atherosclerosis is a chronic artery disease that causes various types of cardiovascular dysfunction. Vascular smooth muscle cells (VSMCs), the main components of atherosclerotic plaque, switch from contractile to synthetic phenotypes during atherogenesis. Ubiquitylation is crucial in regulating VSMC phenotypes in atherosclerosis, and it can be reversely regulated by deubiquitinases. However, the specific effects of deubiquitinases on atherosclerosis have not been thoroughly elucidated. In this study, RNAi screening in human aortic smooth muscle cells was performed to explore the effects of OTU family deubiquitinases, which revealed that silencing OTUB1 inhibited PDGF-BB-stimulated VSMC phenotype switch. Further in vivo studies using Apoe-/- mice revealed that knockdown of OTUB1 in VSMCs alleviated atherosclerosis plaque burden in the advanced stage and led to a stable plaque phenotype. Moreover, VSMC proliferation and migration upon PDGF-BB stimulation could be inhibited by silencing OTUB1 in vitro. Unbiased RNA-sequencing data indicated that knocking down OTUB1 influenced VSMC differentiation, adhesion, and proliferation. Mass spectrometry of ubiquitinated protein confirmed that proteins related to cell growth and migration were differentially ubiquitylated. Mechanistically, we found that OTUB1 recognized the K707 residue ubiquitylation of PDGFRβ with its catalytic triad, thereby reducing the K48-linked ubiquitylation of PDGFRβ. Inhibiting OTUB1 in VSMCs could promote PDGFRβ degradation via the ubiquitin-proteasome pathway, so it was beneficial in preventing VSMCs' phenotype switch. These findings revealed that knocking down OTUB1 ameliorated VSMCs' phenotype switch and atherosclerosis progression, indicating that OTUB1 could be a valuable translational therapeutic target in the future.
Collapse
Affiliation(s)
- Fei Xu
- Department of Cardiology and Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Han Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Changyi Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Tongtong Zang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Rui Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Shutong Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Chaofu Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Yue Yu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Zhiqiang Pei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China.
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Research Unit of Cardiovascular Techniques and Devices, Chinese Academy of Medical Sciences, Shanghai, 200032, China.
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine & Shanghai Clinical Research Center for Interventional Medicine (19MC1910300), Shanghai, 200032, China.
| |
Collapse
|
40
|
Miller LR, Bickel MA, Vance ML, Vaden H, Nagykaldi D, Nyul-Toth A, Bullen EC, Gautam T, Tarantini S, Yabluchanskiy A, Kiss T, Ungvari Z, Conley SM. Vascular smooth muscle cell-specific Igf1r deficiency exacerbates the development of hypertension-induced cerebral microhemorrhages and gait defects. GeroScience 2024; 46:3481-3501. [PMID: 38388918 PMCID: PMC11009188 DOI: 10.1007/s11357-024-01090-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/03/2024] [Indexed: 02/24/2024] Open
Abstract
Cerebrovascular fragility and cerebral microhemorrhages (CMH) contribute to age-related cognitive impairment, mobility defects, and vascular cognitive impairment and dementia, impairing healthspan and reducing quality of life in the elderly. Insulin-like growth factor 1 (IGF-1) is a key vasoprotective growth factor that is reduced during aging. Circulating IGF-1 deficiency leads to the development of CMH and other signs of cerebrovascular dysfunction. Here our goal was to understand the contribution of IGF-1 signaling on vascular smooth muscle cells (VSMCs) to the development of CMH and associated gait defects. We used an inducible VSMC-specific promoter and an IGF-1 receptor (Igf1r) floxed mouse line (Myh11-CreERT2 Igf1rf/f) to knockdown Igf1r. Angiotensin II in combination with L-NAME-induced hypertension was used to elicit CMH. We observed that VSMC-specific Igf1r knockdown mice had accelerated development of CMH, and subsequent associated gait irregularities. These phenotypes were accompanied by upregulation of a cluster of pro-inflammatory genes associated with VSMC maladaptation. Collectively our findings support an essential role for VSMCs as a target for the vasoprotective effects of IGF-1, and suggest that VSMC dysfunction in aging may contribute to the development of CMH.
Collapse
Affiliation(s)
- Lauren R Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
- Currently at: Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Marisa A Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Michaela L Vance
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Hannah Vaden
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Domonkos Nagykaldi
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Adam Nyul-Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Elizabeth C Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA
| | - Tripti Gautam
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Tamas Kiss
- Pediatric Center, Semmelweis University, Budapest, Hungary
- Eötvös Loránd Research Network and Semmelweis University Cerebrovascular and Neurocognitive Disorders Research Group, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon M Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd, BMSB 553, Oklahoma City, OK, 73104, USA.
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
41
|
Chu S, Shan D, He L, Yang S, Feng Y, Zhang Y, Yu J. Anemoside B4 attenuates abdominal aortic aneurysm by limiting smooth muscle cell transdifferentiation and its mediated inflammation. Front Immunol 2024; 15:1412022. [PMID: 38881898 PMCID: PMC11176519 DOI: 10.3389/fimmu.2024.1412022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a degenerative disease characterized by local abnormal dilation of the aorta accompanied by vascular smooth muscle cell (VSMC) dysfunction and chronic inflammation. VSMC dedifferentiation, transdifferentiation, and increased expression of matrix metalloproteinases (MMPs) are essential causes of AAA formation. Previous studies from us and others have shown that Anemoside B4 (AB4), a saponin from Pulsatilla chinensis, has anti-inflammatory, anti-tumor, and regulatory effects on VSMC dedifferentiation. The current study aimed to investigate whether AB4 inhibits AAA development and its underlying mechanisms. By using an Ang II induced AAA model in vivo and cholesterol loading mediated VSMC to macrophage transdifferentiation model in vitro, our study demonstrated that AB4 could attenuate AAA pathogenesis, prevent VSMC dedifferentiation and transdifferentiation to macrophage-like cells, decrease vascular inflammation, and suppress MMP expression and activity. Furthermore, KLF4 overexpression attenuated the effects of AB4 on VSMC to macrophage-like cell transition and VSMC inflammation in vitro. In conclusion, AB4 protects against AAA formation in mice by inhibiting KLF4 mediated VSMC transdifferentiation and inflammation. Our study provides the first proof of concept of using AB4 for AAA management.
Collapse
MESH Headings
- Animals
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/chemically induced
- Cell Transdifferentiation/drug effects
- Kruppel-Like Factor 4/metabolism
- Mice
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Inflammation/metabolism
- Saponins/pharmacology
- Disease Models, Animal
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Mice, Inbred C57BL
- Macrophages/metabolism
- Macrophages/drug effects
- Macrophages/immunology
- Angiotensin II/pharmacology
- Humans
Collapse
Affiliation(s)
- Shuhan Chu
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Dan Shan
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Luling He
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Shilin Yang
- National Pharmaceutical Engineering Center (NPEC) for Solid Preparation in Chinese Herbal Medicine, Nanchang, Jiangxi, China
| | - Yulin Feng
- National Pharmaceutical Engineering Center (NPEC) for Solid Preparation in Chinese Herbal Medicine, Nanchang, Jiangxi, China
| | - Yifeng Zhang
- Center for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Jun Yu
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
42
|
Saul D, Doolittle ML, Rowsey JL, Froemming MN, Kosinsky RL, Vos SJ, Ruan M, LeBrasseur NK, Chandra A, Pignolo RJ, Passos JF, Farr JN, Monroe DG, Khosla S. Osteochondroprogenitor cells and neutrophils expressing p21 and senescence markers modulate fracture repair. J Clin Invest 2024; 134:e179834. [PMID: 38753433 PMCID: PMC11178538 DOI: 10.1172/jci179834] [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: 01/29/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
Cells expressing features of senescence, including upregulation of p21 and p16, appear transiently following tissue injury, yet the properties of these cells or how they contrast with age-induced senescent cells remains unclear. Here, we used skeletal injury as a model and identified the rapid appearance following fracture of p21+ cells expressing senescence markers, mainly as osteochondroprogenitors (OCHs) and neutrophils. Targeted genetic clearance of p21+ cells suppressed senescence-associated signatures within the fracture callus and accelerated fracture healing. By contrast, p21+ cell clearance did not alter bone loss due to aging; conversely, p16+ cell clearance, known to alleviate skeletal aging, did not affect fracture healing. Following fracture, p21+ neutrophils were enriched in signaling pathways known to induce paracrine stromal senescence, while p21+ OCHs were highly enriched in senescence-associated secretory phenotype factors known to impair bone formation. Further analysis revealed an injury-specific stem cell-like OCH subset that was p21+ and highly inflammatory, with a similar inflammatory mesenchymal population (fibro-adipogenic progenitors) evident following muscle injury. Thus, intercommunicating senescent-like neutrophils and mesenchymal progenitor cells were key regulators of tissue repair in bone and potentially across tissues. Moreover, our findings established contextual roles of p21+ versus p16+ senescent/senescent-like cells that may be leveraged for therapeutic opportunities.
Collapse
Affiliation(s)
- Dominik Saul
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Trauma and Reconstructive Surgery, BG Clinic, University of Tübingen, Tübingen, Germany
| | - Madison L. Doolittle
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer L. Rowsey
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Mitchell N. Froemming
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Robyn L. Kosinsky
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Stephanie J. Vos
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Ming Ruan
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physical Medicine and Rehabilitation and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Abhishek Chandra
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert J. Pignolo
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - João F. Passos
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Joshua N. Farr
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - David G. Monroe
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Sundeep Khosla
- Division of Endocrinology and
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
43
|
Xin Y, Zhang Z, Lv S, Xu S, Liu A, Li H, Li P, Han H, Liu Y. Elucidating VSMC phenotypic transition mechanisms to bridge insights into cardiovascular disease implications. Front Cardiovasc Med 2024; 11:1400780. [PMID: 38803664 PMCID: PMC11128571 DOI: 10.3389/fcvm.2024.1400780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/01/2024] [Indexed: 05/29/2024] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, despite advances in understanding cardiovascular health. Significant barriers still exist in effectively preventing and managing these diseases. Vascular smooth muscle cells (VSMCs) are crucial for maintaining vascular integrity and can switch between contractile and synthetic functions in response to stimuli such as hypoxia and inflammation. These transformations play a pivotal role in the progression of cardiovascular diseases, facilitating vascular modifications and disease advancement. This article synthesizes the current understanding of the mechanisms and signaling pathways regulating VSMC phenotypic transitions, highlighting their potential as therapeutic targets in cardiovascular disease interventions.
Collapse
Affiliation(s)
- Yuning Xin
- Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Zipei Zhang
- Traditional Chinese Medicine, The Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Shan Lv
- Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Shan Xu
- Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Aidong Liu
- Traditional Chinese Medicine, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Hongyu Li
- Traditional Chinese Medicine, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Pengfei Li
- Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Huize Han
- Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Yinghui Liu
- Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
44
|
Rojas MG, Pereira-Simon S, Zigmond ZM, Varona Santos J, Perla M, Santos Falcon N, Stoyell-Conti FF, Salama A, Yang X, Long X, Duque JC, Salman LH, Tabbara M, Martinez L, Vazquez-Padron RI. Single-Cell Analyses Offer Insights into the Different Remodeling Programs of Arteries and Veins. Cells 2024; 13:793. [PMID: 38786017 PMCID: PMC11119253 DOI: 10.3390/cells13100793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Arteries and veins develop different types of occlusive diseases and respond differently to injury. The biological reasons for this discrepancy are not well understood, which is a limiting factor for the development of vein-targeted therapies. This study contrasts human peripheral arteries and veins at the single-cell level, with a focus on cell populations with remodeling potential. Upper arm arteries (brachial) and veins (basilic/cephalic) from 30 organ donors were compared using a combination of bulk and single-cell RNA sequencing, proteomics, flow cytometry, and histology. The cellular atlases of six arteries and veins demonstrated a 7.8× higher proportion of contractile smooth muscle cells (SMCs) in arteries and a trend toward more modulated SMCs. In contrast, veins showed a higher abundance of endothelial cells, pericytes, and macrophages, as well as an increasing trend in fibroblasts. Activated fibroblasts had similar proportions in both types of vessels but with significant differences in gene expression. Modulated SMCs and activated fibroblasts were characterized by the upregulation of MYH10, FN1, COL8A1, and ITGA10. Activated fibroblasts also expressed F2R, POSTN, and COMP and were confirmed by F2R/CD90 flow cytometry. Activated fibroblasts from veins were the top producers of collagens among all fibroblast populations from both types of vessels. Venous fibroblasts were also highly angiogenic, proinflammatory, and hyper-responders to reactive oxygen species. Differences in wall structure further explain the significant contribution of fibroblast populations to remodeling in veins. Fibroblasts are almost exclusively located outside the external elastic lamina in arteries, while widely distributed throughout the venous wall. In line with the above, ECM-targeted proteomics confirmed a higher abundance of fibrillar collagens in veins vs. more basement ECM components in arteries. The distinct cellular compositions and transcriptional programs of reparative populations in arteries and veins may explain differences in acute and chronic wall remodeling between vessels. This information may be relevant for the development of antistenotic therapies.
Collapse
Affiliation(s)
- Miguel G. Rojas
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Simone Pereira-Simon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | | | - Javier Varona Santos
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Mikael Perla
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Nieves Santos Falcon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Filipe F. Stoyell-Conti
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Alghidak Salama
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Xiaofeng Yang
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Xiaochun Long
- Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Juan C. Duque
- Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Loay H. Salman
- Division of Nephrology and Hypertension, Albany Medical College, Albany, NY 12208, USA
| | - Marwan Tabbara
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Laisel Martinez
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
| | - Roberto I. Vazquez-Padron
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (M.G.R.); (S.P.-S.); (J.V.S.); (A.S.)
- Bruce W. Carter Veterans Affairs Medical Center, Miami, FL 33125, USA;
| |
Collapse
|
45
|
Yuan X, Jiang C, Xue Y, Guo F, Luo M, Guo L, Gao Y, Yuan T, Xu H, Chen H. KLF13 promotes VSMCs phenotypic dedifferentiation by directly binding to the SM22α promoter. J Cell Physiol 2024; 239:e31251. [PMID: 38634445 DOI: 10.1002/jcp.31251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/24/2024] [Accepted: 02/28/2024] [Indexed: 04/19/2024]
Abstract
Krüppel-like factor 13 (KLF13), a zinc finger transcription factor, is considered as a potential regulator of cardiomyocyte differentiation and proliferation during heart morphogenesis. However, its precise role in the dedifferentiation of vascular smooth muscle cells (VSMCs) during atherosclerosis and neointimal formation after injury remains poorly understood. In this study, we investigated the relationship between KLF13 and SM22α expression in normal and atherosclerotic plaques by bioanalysis, and observed a significant increase in KLF13 levels in the atherosclerotic plaques of both human patients and ApoE-/- mice. Knockdown of KLF13 was found to ameliorate intimal hyperplasia following carotid artery injury. Furthermore, we discovered that KLF13 directly binds to the SM22α promoter, leading to the phenotypic dedifferentiation of VSMCs. Remarkably, we observed a significant inhibition of platelet-derived growth factor BB-induced VSMCs dedifferentiation, proliferation, and migration when knocked down KLF13 in VSMCs. This inhibitory effect of KLF13 knockdown on VCMC function was, at least in part, mediated by the inactivation of p-AKT signaling in VSMCs. Overall, our findings shed light on a potential therapeutic target for treating atherosclerotic lesions and restenosis after vascular injury.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Carotid Artery Injuries/pathology
- Carotid Artery Injuries/genetics
- Carotid Artery Injuries/metabolism
- Cell Dedifferentiation
- Cell Movement/genetics
- Cell Proliferation/genetics
- Cells, Cultured
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Mice, Inbred C57BL
- Muscle Proteins/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neointima/metabolism
- Neointima/pathology
- Neointima/genetics
- Phenotype
- Plaque, Atherosclerotic/pathology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/genetics
- Promoter Regions, Genetic/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Cell Cycle Proteins
- Microfilament Proteins/genetics
Collapse
Affiliation(s)
- Xiaofan Yuan
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Chuan Jiang
- Department of Neurosurgery, The Southwest Medical University, Luzhou, Sichuan, China
| | - Yuzhou Xue
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Fuqiang Guo
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lei Guo
- Department of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yang Gao
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tongling Yuan
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hui Xu
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Hong Chen
- Department of General Practice, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
46
|
Ren H, Hu W, Jiang T, Yao Q, Qi Y, Huang K. Mechanical stress induced mitochondrial dysfunction in cardiovascular diseases: Novel mechanisms and therapeutic targets. Biomed Pharmacother 2024; 174:116545. [PMID: 38603884 DOI: 10.1016/j.biopha.2024.116545] [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: 02/05/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality worldwide. Others and our studies have shown that mechanical stresses (forces) including shear stress and cyclic stretch, occur in various pathological conditions, play significant roles in the development and progression of CVDs. Mitochondria regulate the physiological processes of cardiac and vascular cells mainly through adenosine triphosphate (ATP) production, calcium flux and redox control while promote cell death through electron transport complex (ETC) related cellular stress response. Mounting evidence reveal that mechanical stress-induced mitochondrial dysfunction plays a vital role in the pathogenesis of many CVDs including heart failure and atherosclerosis. This review summarized mitochondrial functions in cardiovascular system under physiological mechanical stress and mitochondrial dysfunction under pathological mechanical stress in CVDs (graphical abstract). The study of mitochondrial dysfunction under mechanical stress can further our understanding of the underlying mechanisms, identify potential therapeutic targets, and aid the development of novel treatments of CVDs.
Collapse
Affiliation(s)
- He Ren
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Weiyi Hu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China
| | - Tao Jiang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Qingping Yao
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China
| | - Yingxin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China
| | - Kai Huang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang, Shanghai 200240, China.
| |
Collapse
|
47
|
Kilanowski-Doroh IM, McNally AB, Wong T, Visniauskas B, Blessinger SA, Sugi AI, Richard C, Diaz Z, Horton A, Natale CA, Ogola BO, Lindsey SH. Ovariectomy-Induced Arterial Stiffening Differs From Vascular Aging and Is Reversed by GPER Activation. Hypertension 2024; 81:e51-e62. [PMID: 38445498 PMCID: PMC11023783 DOI: 10.1161/hypertensionaha.123.22024] [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: 09/07/2023] [Accepted: 02/16/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Arterial stiffness is a cardiovascular risk factor and dramatically increases as women transition through menopause. The current study assessed whether a mouse model of menopause increases arterial stiffness in a similar manner to aging and whether activation of the G-protein-coupled estrogen receptor could reverse stiffness. METHODS Female C57Bl/6J mice were ovariectomized at 10 weeks of age or aged to 52 weeks, and some mice were treated with G-protein-coupled estrogen receptor agonists. RESULTS Ovariectomy and aging increased pulse wave velocity to a similar extent independent of changes in blood pressure. Aging increased carotid wall thickness, while ovariectomy increased material stiffness without altering vascular geometry. RNA-sequencing analysis revealed that ovariectomy downregulated smooth muscle contractile genes. The enantiomerically pure G-protein-coupled estrogen receptor agonist, LNS8801, reversed stiffness in ovariectomy mice to a greater degree than the racemic agonist G-1. In summary, ovariectomy and aging induced arterial stiffening via potentially different mechanisms. Aging was associated with inward remodeling, while ovariectomy-induced material stiffness independent of geometry and a loss of the contractile phenotype. CONCLUSIONS This study enhances our understanding of the impact of estrogen loss on vascular health in a murine model and warrants further studies to examine the ability of LNS8801 to improve vascular health in menopausal women.
Collapse
Affiliation(s)
| | | | - Tristen Wong
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Bruna Visniauskas
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | | | - Chase Richard
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| | - Zaidmara Diaz
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | - Alec Horton
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
| | | | - Benard O. Ogola
- Vascular Biology Center and Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA
| | - Sarah H. Lindsey
- Department of Pharmacology, Tulane School of Medicine, New Orleans, LA
- Tulane Brain Institute, Tulane University, New Orleans, LA
| |
Collapse
|
48
|
Ouyang Y, Hong Y, Mai C, Yang H, Wu Z, Gao X, Zeng W, Deng X, Liu B, Zhang Y, Fu Q, Huang X, Liu J, Li X. Transcriptome analysis reveals therapeutic potential of NAMPT in protecting against abdominal aortic aneurysm in human and mouse. Bioact Mater 2024; 34:17-36. [PMID: 38173843 PMCID: PMC10761368 DOI: 10.1016/j.bioactmat.2023.11.020] [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: 08/20/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
Abdominal Aortic Aneurysm (AAA) is a life-threatening vascular disease characterized by the weakening and ballooning of the abdominal aorta, which has no effective therapeutic approaches due to unclear molecular mechanisms. Using single-cell RNA sequencing, we analyzed the molecular profile of individual cells within control and AAA abdominal aortas. We found cellular heterogeneity, with increased plasmacytoid dendritic cells and reduced endothelial cells and vascular smooth muscle cells (VSMCs) in AAA. Up-regulated genes in AAA were associated with muscle tissue development and apoptosis. Genes controlling VSMCs aberrant switch from contractile to synthetic phenotype were significantly enriched in AAA. Additionally, VSMCs in AAA exhibited cell senescence and impaired oxidative phosphorylation. Similar observations were made in a mouse model of AAA induced by Angiotensin II, further affirming the relevance of our findings to human AAA. The concurrence of gene expression changes between human and mouse highlighted the impairment of oxidative phosphorylation as a potential target for intervention. Nicotinamide phosphoribosyltransferase (NAMPT, also named VISFATIN) signaling emerged as a signature event in AAA. NAMPT was significantly downregulated in AAA. NAMPT-extracellular vesicles (EVs) derived from mesenchymal stem cells restored NAMPT levels, and offered protection against AAA. Furthermore, NAMPT-EVs not only repressed injuries, such as cell senescence and DNA damage, but also rescued impairments of oxidative phosphorylation in both mouse and human AAA models, suggesting NAMPT supplementation as a potential therapeutic approach for AAA treatment. These findings shed light on the cellular heterogeneity and injuries in AAA, and offered promising therapeutic intervention for AAA treatment.
Collapse
Affiliation(s)
- Yu Ouyang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
- Department of Emergency Medicine, The Key Laboratory of Advanced Interdisciplinary Studies , The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yimei Hong
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangdong, 510006, China
| | - Cong Mai
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangdong, 510006, China
| | - Hangzhen Yang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
- Global Health Research Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Zicong Wu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510006, China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangdong, 510006, China
| | - Xiaoyan Gao
- School of Medicine, South China University of Technology, Guangdong, 510006, China
| | - Weiyue Zeng
- School of Medicine, South China University of Technology, Guangdong, 510006, China
| | - Xiaohui Deng
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510006, China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangdong, 510006, China
| | - Baojuan Liu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
| | - Yuelin Zhang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
| | - Qingling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510006, China
- Extracellular Vesicle Research and Clinical Translational Center, The First Affiliated Hospital, Sun Yat-sen University, Guangdong, 510006, China
| | - Xiaojia Huang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Juli Liu
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Xin Li
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangdong, 510006, China
| |
Collapse
|
49
|
Stroope C, Nettersheim FS, Coon B, Finney AC, Schwartz MA, Ley K, Rom O, Yurdagul A. Dysregulated cellular metabolism in atherosclerosis: mediators and therapeutic opportunities. Nat Metab 2024; 6:617-638. [PMID: 38532071 PMCID: PMC11055680 DOI: 10.1038/s42255-024-01015-w] [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: 03/05/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Accumulating evidence over the past decades has revealed an intricate relationship between dysregulation of cellular metabolism and the progression of atherosclerotic cardiovascular disease. However, an integrated understanding of dysregulated cellular metabolism in atherosclerotic cardiovascular disease and its potential value as a therapeutic target is missing. In this Review, we (1) summarize recent advances concerning the role of metabolic dysregulation during atherosclerosis progression in lesional cells, including endothelial cells, vascular smooth muscle cells, macrophages and T cells; (2) explore the complexity of metabolic cross-talk between these lesional cells; (3) highlight emerging technologies that promise to illuminate unknown aspects of metabolism in atherosclerosis; and (4) suggest strategies for targeting these underexplored metabolic alterations to mitigate atherosclerosis progression and stabilize rupture-prone atheromas with a potential new generation of cardiovascular therapeutics.
Collapse
Affiliation(s)
- Chad Stroope
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Felix Sebastian Nettersheim
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Brian Coon
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Cardiovascular Biology Research Program, OMRF, Oklahoma City, OK, USA
- Department of Cell Biology, Oklahoma University Health Sciences Center, Oklahoma City, OK, USA
| | - Alexandra C Finney
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Martin A Schwartz
- Yale Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Departments of Cell Biology and Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Klaus Ley
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
- Immunology Center of Georgia (IMMCG), Augusta University Immunology Center of Georgia, Augusta, GA, USA
| | - Oren Rom
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, USA.
| |
Collapse
|
50
|
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
|