1
|
Dou Y, Zhang X, Guo R, Huang X, Song Y, Liu X, Shi J, Li F, Zhang D, Kong P, Nie L, Li H, Zhang F, Han M. Quaking-cZFP609 Axis Remedies Aberrant Plasticity of Vascular Smooth Muscle Cells via Mediating Platelet-Derived Growth Factor Receptor β Degradation. MedComm (Beijing) 2025; 6:e70167. [PMID: 40242156 PMCID: PMC12000678 DOI: 10.1002/mco2.70167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 04/18/2025] Open
Abstract
Vascular smooth muscle cell (VSMC) plasticity is crucial for the repair after vascular injury. However, the high plasticity of VSMCs may make them transform into pathogenic phenotypes. Here, we show that VSMCs overexpressing Sirtuin 1 (SIRT1) exhibit a reduced phenotypic plasticity in the context of platelet-derived growth factor (PDGF)-BB treatment. SIRT1 activated Quaking (QKI)-cZFP609 axis is involved in the plasticity regulation in the VSMCs. Mechanically, SIRT1 deacetylates K133 and K134 of QKI and mediates its activation. Activated QKI binds the QKI response elements located in the upstream and downstream of the cZFP609-forming exons in ZFP609 pre-mRNA to mediate cZFP609 production. Furthermore, the acetylation of QKI is increased by inhibiting SIRT1 with the selective and potent inhibitor EX527 or deletion of SIRT1, accompanied with parallel decrease in cZFP609 formation. Final, we identify that cZFP609 directs PDGF receptor (PDGFR)β sorting into endosomal/lysosomal pathway and degradation by bridging PDGFRβ and Rab7, resulted in attenuating Raf-MEK-ERK cascade activation downstream of PDGFRβ signaling. Overexpression of cZFP609 remedies aberrant plasticity and overproliferation of VSMCs, and ameliorates neointimal formation. Together, these results highlight that modulating the QKI-cZFP609 axis may help propel repair without stenosis as a therapeutic strategy in vascular injury.
Collapse
Affiliation(s)
- Yong‐Qing Dou
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- College of Integrative MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Xiao‐Yun Zhang
- College of Integrative MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Rui‐Juan Guo
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| | - Xiao‐Fu Huang
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
| | - Yu Song
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| | - Xin‐Long Liu
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
| | - Jie Shi
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
| | - Fan‐Qin Li
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
| | - Dan‐Dan Zhang
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| | - Peng Kong
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| | - Lei Nie
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| | - Han Li
- Department of Orthopaedic SurgeryInstitute of Biomechanical Science and Biomechanical Key Laboratory of Hebei ProvinceThird Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Fan Zhang
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| | - Mei Han
- Department of Biochemistry and Molecular BiologyCollege of Basic MedicineShijiazhuangChina
- Key Laboratory of Neural and Vascular Biology of Ministry of EducationShijiazhuangChina
- Key Laboratory of Vascular Biology of Hebei ProvinceHebei Medical UniversityShijiazhuangChina
| |
Collapse
|
2
|
Bolanle IO, Palmer TM. O-GlcNAcylation and Phosphorylation Crosstalk in Vascular Smooth Muscle Cells: Cellular and Therapeutic Significance in Cardiac and Vascular Pathologies. Int J Mol Sci 2025; 26:3303. [PMID: 40244145 PMCID: PMC11989994 DOI: 10.3390/ijms26073303] [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: 01/27/2025] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
More than 400 different types of post-translational modifications (PTMs), including O-GlcNAcylation and phosphorylation, combine to co-ordinate almost all aspects of protein function. Often, these PTMs overlap and the specific relationship between O-GlcNAcylation and phosphorylation has drawn much attention. In the last decade, the significance of this dynamic crosstalk has been linked to several chronic pathologies of cardiovascular origin. However, very little is known about the pathophysiological significance of this crosstalk for vascular smooth muscle cell dysfunction in cardiovascular disease. O-GlcNAcylation occurs on serine and threonine residues which are also targets for phosphorylation. A growing body of research has now emerged linking altered vascular integrity and homeostasis with highly regulated crosstalk between these PTMs. Additionally, a significant body of evidence indicates that O-GlcNAcylation is an important contributor to the pathogenesis of neointimal hyperplasia and vascular restenosis responsible for long-term vein graft failure. In this review, we evaluate the significance of this dynamic crosstalk and its role in cardiovascular pathologies, and the prospects of identifying possible targets for more effective therapeutic interventions.
Collapse
Affiliation(s)
| | - Timothy M. Palmer
- Biomedical Institute for Multimorbidity, Centre for Biomedicine, Hull York Medical School, University of Hull, Hull HU6 7RX, UK;
| |
Collapse
|
3
|
Newman AAC, Dalman JM, Moore KJ. Cardiovascular Disease and Cancer: A Dangerous Liaison. Arterioscler Thromb Vasc Biol 2025; 45:359-371. [PMID: 39781742 PMCID: PMC11864891 DOI: 10.1161/atvbaha.124.319863] [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: 11/08/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The field of cardio-oncology has traditionally focused on the impact of cancer and its therapies on cardiovascular health. Mounting clinical and preclinical evidence, however, indicates that the reverse may also be true: cardiovascular disease can itself influence tumor growth and metastasis. Numerous epidemiological studies have reported that individuals with prevalent cardiovascular disease have an increased incidence of cancer. In parallel, studies using preclinical mouse models of myocardial infarction, heart failure, and cardiac remodeling support the notion that cardiovascular disorders accelerate the growth of solid tumors and metastases. These findings have ushered in a new and burgeoning field termed reverse cardio-oncology that investigates the impact of cardiovascular disease pathophysiology on cancer emergence and progression. Recent studies have begun to illuminate the mechanisms driving this relationship, including shared risk factors, reprogramming of immune responses, changes in gene expression, and the release of cardiac factors that result in selective advantages for tumor cells or their local milieu, thus exacerbating cancer pathology. Here, we review the evidence supporting the relationship between cardiovascular disease and cancer, the mechanistic pathways enabling this connection, and the implications of these findings for patient care.
Collapse
Affiliation(s)
- Alexandra A C Newman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jessie M Dalman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- University of Michigan Medical School, Ann Arbor, MI 48104, USA
| | - Kathryn J Moore
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
4
|
Tan JM, Cheng L, Calhoun RP, Weller AH, Drareni K, Fong S, Barbara E, Lim HW, Xue C, Winter H, Auguste G, Miller CL, Reilly MP, Maegdefessel L, Lutgens E, Seale P. PRDM16 controls smooth muscle cell fate in atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639186. [PMID: 40027729 PMCID: PMC11870537 DOI: 10.1101/2025.02.19.639186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Vascular smooth muscle cells (SMCs) normally exist in a contractile state but can undergo fate switching to produce various cell phenotypes in response to pathologic stimuli 1-3 . In atherosclerosis, these phenotypically modulated SMCs regulate plaque composition and influence the risk of major adverse cardiovascular events 4,5 . We found that PRDM16, a transcription factor that is genetically associated with cardiovascular disease, is highly expressed in arterial SMCs and downregulated during SMC fate switching in human and mouse atherosclerosis. Loss of Prdm16 in SMCs of mice activates a synthetic modulation program under homeostatic conditions. Single cell analyses show that loss of Prdm16 drives a synthetic program in all SMC populations. Upon exposure to atherogenic stimuli, SMC-selective Prdm16 deficient mice develop SMC-rich, fibroproliferative plaques that contain few foam cells. Acute loss of Prdm16 results in the formation of collagen-rich lesions with thick fibrous caps. Reciprocally, increasing PRDM16 expression in SMCs blocks synthetic processes, including migration, proliferation, and fibrosis. Mechanistically, PRDM16 binds to chromatin and decreases activating histone marks at synthetic genes. Altogether, these results define PRDM16 as a critical determinant of SMC identity and atherosclerotic lesion composition.
Collapse
Affiliation(s)
- Josephine M.E. Tan
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lan Cheng
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan P. Calhoun
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela H. Weller
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karima Drareni
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Skylar Fong
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eirlys Barbara
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hee-Woong Lim
- Department of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, OH, USA
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| | - Hanna Winter
- Institute of Molecular Vascular Medicine, University Hospital rechts der Isar, Technical University Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany
| | - Gaëlle Auguste
- Department of Genome Sciences; Department of Biochemistry and Molecular Genetics; University of Virginia, Charlottesville, VA 22908, USA
| | - Clint L. Miller
- Department of Genome Sciences; Department of Biochemistry and Molecular Genetics; University of Virginia, Charlottesville, VA 22908, USA
| | - Muredach P. Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA
| | - Lars Maegdefessel
- Institute of Molecular Vascular Medicine, University Hospital rechts der Isar, Technical University Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Germany
| | - Esther Lutgens
- Experimental Cardiovascular Immunology Laboratory, Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity & Metabolism; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology; Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
5
|
Lambert J, Jørgensen HF. Epigenetic regulation of vascular smooth muscle cell phenotypes in atherosclerosis. Atherosclerosis 2025; 401:119085. [PMID: 39709233 DOI: 10.1016/j.atherosclerosis.2024.119085] [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: 07/16/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
Vascular smooth muscle cells (VSMCs) in adult arteries maintain substantial phenotypic plasticity, which allows for the reversible cell state changes that enable vascular remodelling and homeostasis. In atherosclerosis, VSMCs dedifferentiate in response to lipid accumulation and inflammation, resulting in loss of their characteristic contractile state. Recent studies showed that individual, pre-existing VSMCs expand clonally and can acquire many different phenotypes in atherosclerotic lesions. The changes in gene expression underlying this phenotypic diversity are mediated by epigenetic modifications which affect transcription factor access and thereby gene expression dynamics. Additionally, epigenetic mechanisms can maintain cellular memory, potentially facilitating reversion to the contractile state. While technological advances have provided some insight, a comprehensive understanding of how VSMC phenotypes are governed in disease remains elusive. Here we review current literature in light of novel insight from studies at single-cell resolution. We also discuss how lessons from epigenetic studies of cellular regulation in other fields could help in translating the potential of targeting VSMC phenotype conversion into novel therapies in cardiovascular disease.
Collapse
Affiliation(s)
- Jordi Lambert
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| | - Helle F Jørgensen
- Section of Cardiorespiratory Medicine, University of Cambridge, VPD Heart and Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, Cambridge, CB2 0BB, UK.
| |
Collapse
|
6
|
Maegdefessel L, Fasolo F. Long Non-Coding RNA Function in Smooth Muscle Cell Plasticity and Atherosclerosis. Arterioscler Thromb Vasc Biol 2025; 45:172-185. [PMID: 39633574 PMCID: PMC11748911 DOI: 10.1161/atvbaha.124.320393] [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: 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 non-coding 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 non-coding RNA function with unprecedented precision. Here, we discuss the state of the art of long non-coding RNAs regulation of SMC phenotypic switching, describing the methodologies used to approach this issue and evaluating the therapeutic perspectives of exploiting long non-coding 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
|
7
|
Summer S, Borrell-Pages M, Bruno RM, Climie RE, Dipla K, Dogan A, Eruslanova K, Fraenkel E, Mattace-Raso F, Pugh CJA, Rochfort KD, Ross M, Roth L, Schmidt-Trucksäss A, Schwarz D, Shadiow J, Sohrabi Y, Sonnenberg J, Tura-Ceide O, Guvenc Tuna B, Julve J, Dogan S. Centenarians-the way to healthy vascular ageing and longevity: a review from VascAgeNet. GeroScience 2025; 47:685-702. [PMID: 39725804 PMCID: PMC11872877 DOI: 10.1007/s11357-024-01467-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
The prevalence of centenarians, people who lived 100 years and longer, is steadily growing in the last decades. This exceptional longevity is based on multifaceted processes influenced by a combination of intrinsic and extrinsic factors such as sex, (epi-)genetic factors, gut microbiota, cellular metabolism, exposure to oxidative stress, immune status, cardiovascular risk factors, environmental factors, and lifestyle behavior. Epidemiologically, the incidence rate of cardiovascular diseases is reduced in healthy centenarians along with late onset of age-related diseases compared with the general aged population. Understanding the mechanisms that affect vascular ageing in centenarians and the underlying factors could offer valuable insights for developing strategies to improve overall healthy life span in the elderly. This review discusses these key factors influencing vascular ageing and how their modulation could foster healthy longevity.
Collapse
Affiliation(s)
- Sabrina Summer
- Department for Biomedical Research, University for Continuing Education Krems, Krems, Austria
| | - Maria Borrell-Pages
- Molecular Pathology and Therapeutic of Ischemic and Atherothrombotic Diseases, Institute de Recerca Sant Pau (IR-Sant Pau), Barcelona Spain. CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa-Maria Bruno
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center-PARCC, Paris, France
- Clinical Pharmacology Unit, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Rachel E Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Konstantina Dipla
- Department of Sports Sciences at Serres, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aysenur Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Kseniia Eruslanova
- Russian Gerontology Research and Clinical Centre, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Emil Fraenkel
- Department of Internal Medicine, University of Košice, Košice, Slovakia
| | | | | | - Keith D Rochfort
- School of Nursing, Psychotherapy and Community Health, Dublin City University, Dublin, Ireland
| | - Mark Ross
- Institute of Life and Earth Sciences, Heriot-Watt University, Edinburgh, UK
| | - Lynn Roth
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Arno Schmidt-Trucksäss
- Department of Sport, Exercise and Health, Division of Sport and Exercise Medicine, University of Basel, Basel, Switzerland
| | - Dennis Schwarz
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, University of Münster, Münster, Germany
| | - James Shadiow
- School of Kinesiology, University of Michigan, Ann Arbor, MI, USA
| | - Yahya Sohrabi
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, University of Münster, Münster, Germany
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Jannik Sonnenberg
- Department of Cardiology I-Coronary and Peripheral Vascular Disease, University of Münster, Münster, Germany
| | | | - Bilge Guvenc Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Josep Julve
- Endocrinology, Diabetes and Nutrition Group, Institut de Recerca SANT PAU, Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Türkiye.
| |
Collapse
|
8
|
Pervaiz N, Mehmood R, Aithabathula RV, Kathuria I, Ahn W, Le BT, Kim KS, Singh UP, Csanyi G, Singla B. Smooth muscle cell-specific CD47 deletion suppresses atherosclerosis. Life Sci 2025; 361:123315. [PMID: 39675550 PMCID: PMC11740882 DOI: 10.1016/j.lfs.2024.123315] [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: 09/09/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Recent smooth muscle cell (SMC)-lineage tracing and single-cell RNA sequencing (scRNA-seq) experiments revealed a significant role of SMC-derived cells in atherosclerosis development. Further, thrombospondin-1 (TSP1), a matricellular protein, and activation of its receptor cluster of differentiation (CD) 47 have been linked with atherosclerosis. However, the role of vascular SMC TSP1-CD47 signaling in regulating VSMC phenotype and atherogenesis remains unknown. METHODS We investigated the role of SMC CD47 activation by TSP1 in regulating VSMC phenotype and atherosclerosis development using various in vitro cell-based assays, molecular biological techniques, immunohistological approaches, reanalysis of publicly available scRNA-seq data, and cell-specific knockout mice. RESULTS We observed elevated TSP1 expression in human atherosclerotic vascular tissues and VSMCs. TSP1-treated VSMCs exhibited decreased expression of contractile SMC markers (ACTA2, CNN1, and TAGLN) and increased proliferation. Additional experiments and reanalysis of the scRNA-seq dataset showed CD47 as the major TSP1 receptor in VSMCs, with its expression increased in SMC-derived modulated cells of murine atherosclerotic arteries. Knockdown of CD47 gene in human VSMCs upregulated expression of contractile SMC markers and abrogated TSP1's effects on these genes. SMC-specific Cd47 deletion in mice suppressed atherosclerotic lesion formation, reduced macrophage accumulation, and decreased necrotic area. However, no significant differences were observed in weight gain, liver and adipose tissue mass, plasma total cholesterol, and fasting blood glucose between control and SMC-restricted Cd47-deficient mice. Further experiments demonstrated increased efferocytosis of apoptotic CD47-silenced VSMCs by macrophages. CONCLUSIONS These findings suggest that CD47 plays a crucial role in regulating VSMC phenotype, and SMC-specific-Cd47 deletion suppresses atherosclerosis. NEW AND NOTEWORTHY VSMC phenotypic switching contributes to atherosclerosis development. The present study reports the novel observations that Cd47 levels are upregulated in phenotypically modulated SMCs within atherosclerotic arteries and targeted deletion of Cd47 specifically in SMCs attenuates atherosclerosis. Mechanistic in vitro investigations further showed that TSP1-CD47 signaling regulates VSMC phenotype. Therefore, targeting SMC CD47 represents a promising therapeutic target to suppress atherogenesis.
Collapse
Affiliation(s)
- Naveed Pervaiz
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Rashid Mehmood
- Department of Hematology, St. Jude Children's Hospital, Memphis, TN, USA
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ishita Kathuria
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - WonMo Ahn
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Britney-Thuy Le
- Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ki-Suk Kim
- Department of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Udai P Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA
| | - Gabor Csanyi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Bhupesh Singla
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
9
|
Lehners M, Schmidt H, Zaldivia MTK, Stehle D, Krämer M, Peter A, Adler J, Lukowski R, Feil S, Feil R. Single-cell analysis identifies the CNP/GC-B/cGMP axis as marker and regulator of modulated VSMCs in atherosclerosis. Nat Commun 2025; 16:429. [PMID: 39814746 PMCID: PMC11735800 DOI: 10.1038/s41467-024-55687-9] [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: 03/25/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025] Open
Abstract
A balanced activity of cGMP signaling contributes to the maintenance of cardiovascular homeostasis. Vascular smooth muscle cells (VSMCs) can generate cGMP via three ligand-activated guanylyl cyclases, the NO-sensitive guanylyl cyclase, the atrial natriuretic peptide (ANP)-activated GC-A, and the C-type natriuretic peptide (CNP)-stimulated GC-B. Here, we study natriuretic peptide signaling in murine VSMCs and atherosclerotic lesions. Correlative profiling of pathway activity and VSMC phenotype at the single-cell level shows that phenotypic modulation of contractile VSMCs to chondrocyte-like plaque cells during atherogenesis is associated with a switch from ANP/GC‑A to CNP/GC‑B signaling. Silencing of the CNP/GC-B axis in VSMCs results in an increase of chondrocyte-like plaque cells. These findings indicate that the CNP/GC-B/cGMP pathway is a marker and atheroprotective regulator of modulated VSMCs, limiting their transition to chondrocyte-like cells. Overall, this study highlights the plasticity of cGMP signaling in VSMCs and suggests analogies between CNP-dependent remodeling of bone and blood vessels.
Collapse
Affiliation(s)
- Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Hannes Schmidt
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Maria T K Zaldivia
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Michael Krämer
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Andreas Peter
- Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Julia Adler
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
10
|
Zhao C, Shen J, Lu Y, Ni H, Xiang M, Xie Y. Dedifferentiation of vascular smooth muscle cells upon vessel injury. Int Immunopharmacol 2025; 144:113691. [PMID: 39591824 DOI: 10.1016/j.intimp.2024.113691] [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: 07/10/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024]
Abstract
Highly differentiated mature vascular smooth muscle cells (VSMCs) are the predominant type of cells constituting arterial walls, which are essential for maintaining the structural and functional integrity of blood vessels. VSMCs demonstrate a notable degree of adaptability following vascular damage, a characteristic that plays a crucial role in the progression of vascular remodeling. Advances in single-cell RNA sequencing in both healthy and pathological vascular tissues have offered profound insights into the complexity of VSMCs, revealing a more intricate diversity than previously recognized. In response to injury, VSMCs undergo dedifferentiation and exhibit pluripotent markers. This review summarizes the researches that have employed lineage tracing alongside single-cell sequencing analysis to explore the dynamics of vascular damage. The primary focus of this study was on the process of dedifferentiation in VSMCs, with particular attention to its underlying mechanisms. The discussion included the impact of microenvironmental cues, the control of transcription factors, and the various molecular pathways involved in VSMCs dedifferentiation. Herein, we provide a comprehensive analysis of cells dedifferentiated from adult VSMCs upon vascular injury.
Collapse
Affiliation(s)
- Chaoyue Zhao
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Transvascular Implantation Devices, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Transvascular Implantation Devices, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Yunrui Lu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Transvascular Implantation Devices, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Transvascular Implantation Devices, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Transvascular Implantation Devices, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Transvascular Implantation Devices, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
11
|
Cheng X, Li YL, Wang H, Zhang RJ, Fan KY, Qi XT, Zheng GP, Dong HL. Mesenchymal stem cell therapy in atherosclerosis: A bibliometric and visual analysis. World J Stem Cells 2024; 16:1062-1085. [PMID: 39734478 PMCID: PMC11669984 DOI: 10.4252/wjsc.v16.i12.1062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/15/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are capable of self-renewal and differentiation, and extensive studies have demonstrated their therapeutic potential in atherosclerosis (AS). AIM To conduct a bibliometric analysis of studies on the use of MSC therapy for AS over the past two decades, assess key trends and provide insights for future research directions. METHODS We systematically searched the Web of Science Core Collection database for articles published between 1999 and 2023, yielding a total of 556 articles. Visual representation and bibliometric analysis of information and trends were facilitated using CiteSpace, the R package 'bibliometrix' and VOSviewer. RESULTS The analyzed articles were predominantly from 52 countries/regions, with prominent contributions from China and the United States. A cohort of 3057 authors contributed to these publications, with the works of Libby P distinguished by their influence and citation count. Int J Mol Sci has emerged as the journal with the highest publication volume, prominently disseminating influential papers and identifying citation outbreaks. Furthermore, our analysis identified current research hotspots within the field, focusing on vascular progenitor cells, inflammatory mechanisms, and extracellular vesicles. Emerging research frontiers, such as extracellular vesicles and oxidative stress, have been highlighted as areas of burgeoning interest. Finally, we offer perspectives on the status of research and future directions of MSC therapy in AS. CONCLUSION This comprehensive analysis provides valuable insights for advancing scientific research on MSC therapy for AS. By elucidating pivotal trends and research directions, this study aimed to foster innovation and promote the progress of disciplines in this field, thereby contributing to advancing scientific knowledge and clinical practice.
Collapse
Affiliation(s)
- Xing Cheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
| | - Ya-Ling Li
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
| | - Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney 2145, New South Wales, Australia
| | - Rui-Jing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
| | - Ke-Yi Fan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
| | - Xiao-Tong Qi
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
| | - Guo-Ping Zheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney 2145, New South Wales, Australia
| | - Hong-Lin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan 030000, Shanxi Province, China.
| |
Collapse
|
12
|
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
|
13
|
Larsen JH, Hegelund JS, Pedersen MK, Andersson CM, Lindegaard CA, Hansen DR, Stubbe J, Lindholt JS, Hansen CS, Grentzmann A, Bloksgaard M, Jensen BL, Rodriguez-Díez RR, Ruiz-Ortega M, Albinsson S, Pasterkamp G, Mokry M, Leask A, Goldschmeding R, Pilecki B, Sorensen GL, Pyke C, Overgaard M, Beck HC, Ketelhuth DFJ, Rasmussen LM, Steffensen LB. Smooth muscle-specific deletion of cellular communication network factor 2 causes severe aorta malformation and atherosclerosis. Cardiovasc Res 2024; 120:1851-1868. [PMID: 39167826 PMCID: PMC11630017 DOI: 10.1093/cvr/cvae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 04/10/2024] [Accepted: 06/22/2024] [Indexed: 08/23/2024] Open
Abstract
AIMS Cellular communication network factor 2 (CCN2) is a matricellular protein implicated in fibrotic diseases, with ongoing clinical trials evaluating anti-CCN2-based therapies. By uncovering CCN2 as abundantly expressed in non-diseased artery tissue, this study aimed to investigate the hypothesis that CCN2 plays a pivotal role in maintaining smooth muscle cell (SMC) phenotype and protection against atherosclerosis. METHODS AND RESULTS Global- and SMC-specific Ccn2 knockout mouse models were employed to demonstrate that Ccn2 deficiency leads to SMC de-differentiation, medial thickening, and aorta elongation under normolipidaemic conditions. Inducing hyperlipidaemia in both models resulted in severe aorta malformation and a 17-fold increase in atherosclerosis formation. Lipid-rich lesions developed at sites of the vasculature typically protected from atherosclerosis development by laminar blood flow, covering 90% of aortas and extending to other vessels, including coronary arteries. Evaluation at earlier time points revealed medial lipid accumulation as a lesion-initiating event. Fluorescently labelled LDL injection followed by confocal microscopy showed increased LDL retention in the medial layer of Ccn2 knockout aortas, likely attributed to marked proteoglycan enrichment of the medial extracellular matrix. Analyses leveraging data from the Athero-Express study cohort indicated the relevance of CCN2 in established human lesions, as CCN2 correlated with SMC marker transcripts across 654 transcriptomically profiled carotid plaques. These findings were substantiated through in situ hybridization showing CCN2 expression predominantly in the fibrous cap. CONCLUSION This study identifies CCN2 as a major constituent of the normal artery wall, critical in regulating SMC differentiation and aorta integrity and possessing a protective role against atherosclerosis development. These findings underscore the need for further investigation into the potential effects of anti-CCN2-based therapies on the vasculature.
Collapse
MESH Headings
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Aorta/metabolism
- Aorta/pathology
- Disease Models, Animal
- Connective Tissue Growth Factor/metabolism
- Connective Tissue Growth Factor/genetics
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Humans
- Phenotype
- Aortic Diseases/genetics
- Aortic Diseases/pathology
- Aortic Diseases/metabolism
- Aortic Diseases/prevention & control
- Vascular Malformations/genetics
- Vascular Malformations/metabolism
- Vascular Malformations/pathology
- Plaque, Atherosclerotic
- Mice, Inbred C57BL
- Genetic Predisposition to Disease
- Male
- Signal Transduction
- Lipoproteins, LDL/metabolism
Collapse
Affiliation(s)
- Jannik H Larsen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
- Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, J. B. Winsløws Vej 4, DK-5000 Odense C, Denmark
| | - Julie S Hegelund
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Matilde K Pedersen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Cecilie M Andersson
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Caroline A Lindegaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Didde R Hansen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Jane Stubbe
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Jes S Lindholt
- Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, J. B. Winsløws Vej 4, DK-5000 Odense C, Denmark
- Department of Cardiothoracic and Vascular Surgery, Odense University Hospital, Odense, Denmark
| | - Camilla S Hansen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Andrietta Grentzmann
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Maria Bloksgaard
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Boye L Jensen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Raúl R Rodriguez-Díez
- Department of Cell Biology, Complutense University School of Medicine, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Sebastian Albinsson
- Department of Experimental Medical Science, Vascular Physiology Environment, Lund University, Lund, Sweden
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, The Netherlands
| | - Michal Mokry
- Laboratory of Clinical Chemistry and Haematology, University Medical Center, Heidelberglaan 100, Utrecht, The Netherlands
- Department of Cardiology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, The Netherlands
| | - Andrew Leask
- College of Dentistry, University of Saskatoon, Saskatoon, SK, Canada
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bartosz Pilecki
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Grith L Sorensen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Charles Pyke
- Research and Early Development, Novo Nordisk A/S, Måløv, Denmark
| | - Martin Overgaard
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Hans C Beck
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Daniel F J Ketelhuth
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Lars M Rasmussen
- Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, J. B. Winsløws Vej 4, DK-5000 Odense C, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Lasse B Steffensen
- Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
- Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital, J. B. Winsløws Vej 4, DK-5000 Odense C, Denmark
| |
Collapse
|
14
|
Lareyre F, Clément M, Cong Duy L, Raffort J. Reply to "Paeonol Suppresses Vasculogenesis Through Regulating Vascular Smooth Muscle Phenotypic Switching". J Endovasc Ther 2024; 31:1268-1269. [PMID: 36927136 DOI: 10.1177/15266028231161233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Affiliation(s)
- Fabien Lareyre
- Department of Vascular Surgery, Hospital of Antibes-Juan-les-Pins, Antibes, France
- Université Côte d'Azur, Inserm U1065, C3M, Nice, France
| | | | - Lê Cong Duy
- Department of Vascular Surgery, Hospital of Antibes-Juan-les-Pins, Antibes, France
- Université Côte d'Azur, Inserm U1065, C3M, Nice, France
| | - Juliette Raffort
- Université Côte d'Azur, Inserm U1065, C3M, Nice, France
- Clinical Chemistry Laboratory, University Hospital of Nice, Nice, France
| |
Collapse
|
15
|
Pløen GG, Sørensen CB, Bentzon JF. Smooth muscle cells clonally expand in a murine carotid allograft model complicated by immune reactions to reporter transgenes. Transpl Immunol 2024; 87:102129. [PMID: 39260676 DOI: 10.1016/j.trim.2024.102129] [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: 07/05/2024] [Revised: 08/27/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND AND AIMS Most experimental studies of allograft vasculopathy (AV) have relied on transplantation between major histocompatibility complex-mismatched inbred mouse strains, but this leads to the complete eradication of donor smooth muscle cells (SMCs) and lesions formed by recipient cells. This is unlike human AV which is thought to form mainly by donor SMCs. Here, we studied sources of neointimal cells in a minor histocompatibility antigen-mismatched AV model by combining male-to-female orthotopic carotid transplantations and lineage tracing by SMC-specific expression of fluorescent proteins. METHODS To track SMC-derived cells in allograft vasculopathy, we used male donor mice with SMC-restricted Cre recombination of the mT/mG reporter transgene, which switches expression of membrane-bound red fluorescent protein (RFP) to green fluorescent protein (GFP), or the stochastically recombining Confetti reporter transgene, which yields a mosaic expression of four fluorescent proteins. Donor carotid segments were harvested and orthotopically allografted to female recipients that were wildtype or had non-recombined reporter transgenes. Inhibition of T cell responses by CTLA4Ig was used in some experiments. Sections of lesions harvested after 4 weeks were analyzed by fluorescence microscopy. RESULTS Donor-derived SMCs survived and gave rise to part of the neointimal cells in experiments where carotid segments from recombined mT/mG male mice were transplanted into wild-type or non-recombined mT/mG female mice. Sex-mismatched transplants developed significant lesions, increasing the intimal and medial area 4.6-fold (p = 0.038) and 2.0-fold (p = 0.024) compared to sex- and fluorescence-matched controls, respectively. Interestingly, sex-matched fluorescence-positive transplants developed intimal lesions in 50% of fluorescence-naïve recipient controls. To study the clonal structure of the neointimal donor-derived SMC lineage cells, we then transplanted male carotids with heterozygous or homozygous recombined Confetti transgenes into female recipients. These transplants developed lesions with few surviving donor SMCs, indicating that expression of the Confetti reporter increased rejection and donor-specific SMC death. Some of the few remaining donor SMCs underwent clonal expansion. CTLA4Ig administration at the time of surgery did not improve SMC survival in mT/mG or Confetti transplants. CONCLUSION Male-to-female transplant models feature donor-derived SMCs, some of which undergo clonal expansion, but immune rejection to fluorescence reporters appears to bias results in lineage tracing models. Overcoming these challenges with alternative reporter transgenes or tolerant recipients is necessary to study the mechanisms by which donor SMCs contribute to allograft vasculopathy.
Collapse
Affiliation(s)
| | | | - Jacob Fog Bentzon
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| |
Collapse
|
16
|
Du D, Bhardwaj S, Lu Y, Wang Y, Parker SJ, Zhang Z, Van Eyk JE, Yu G, Clarke R, Herrington DM, Wang Y. Embracing the informative missingness and silent gene in analyzing biologically diverse samples. Sci Rep 2024; 14:28265. [PMID: 39550430 PMCID: PMC11569126 DOI: 10.1038/s41598-024-78076-0] [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: 05/14/2024] [Accepted: 10/28/2024] [Indexed: 11/18/2024] Open
Abstract
Bioinformatics software tools are essential to identify informative molecular features that define different phenotypic sample groups. Among the most fundamental and interrelated tasks are missing value imputation, signature gene detection, and differential pattern visualization. However, many commonly used analytics tools can be problematic when handling biologically diverse samples if either informative missingness possess high missing rates with mixed missing mechanisms, or multiple sample groups are compared and visualized in parallel. We developed the ABDS tool suite specifically for analyzing biologically diverse samples. Collectively, a mechanism-integrated group-wise pre-imputation scheme is proposed to retain informative missingness associated with signature genes, a cosine-based one-sample test is extended to detect group-silenced signature genes, and a unified heatmap is designed to display multiple sample groups. We describe the methodological principles and demonstrate the effectiveness of three analytics tools under targeted scenarios, supported by comparative evaluations and biomedical showcases. As an open-source R package, ABDS tool suite complements rather than replaces existing tools and will allow biologists to more accurately detect interpretable molecular signals among phenotypically diverse sample groups.
Collapse
Affiliation(s)
- Dongping Du
- Department of Electrical & Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Saurabh Bhardwaj
- Department of Electrical & Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
- Department of Electrical and Instrumentation Engineering, Thapar Institute of Engineering and Technology, Patiala, 147004, Punjab, India
| | - Yingzhou Lu
- Department of Electrical & Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Yizhi Wang
- Department of Electrical & Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA
| | - Sarah J Parker
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Zhen Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, MD, 21231, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing, 100084, P. R. China
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - David M Herrington
- Department of Internal Medicine, Wake Forest University, Winston-Salem, NC, 27157, USA
| | - Yue Wang
- Department of Electrical & Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA, 22203, USA.
- Dept. of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, 900 N. Glebe Road, Arlington, VA, 22203, USA.
| |
Collapse
|
17
|
Guo J, Du L. An update on ox-LDL-inducing vascular smooth muscle cell-derived foam cells in atherosclerosis. Front Cell Dev Biol 2024; 12:1481505. [PMID: 39524227 PMCID: PMC11543427 DOI: 10.3389/fcell.2024.1481505] [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: 08/16/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Excess cholesterol accumulation induces the accumulation of foam cells, eventually accelerating atherosclerosis progress. Historically, the mechanisms of macrophage-derived foam cells have attracted attention because of their central role in plaque development, which was challenged by lineage tracing in union with single-cell sequencing (sc-seq). Accumulated studies have uncovered how vascular smooth muscle cells (VSMCs) proliferate and migrate to the vascular intima and accumulate, then transform into foam cells induced by surplus lipids, finally accounting for 30% to 70% of the total foam cells within the plaque of both mice and humans. Therefore, the mechanisms of VSMC-derived foam cells have received increasing attention. The review intends to summarize the transformation mechanism of VSMCs into foam cells induced by oxidized low-density lipoproteins (ox-LDL) in atherosclerosis.
Collapse
Affiliation(s)
- Jingjing Guo
- Luoyang Key Laboratory of Cardiovascular Science, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Laijing Du
- Department of Cardiology, Henan Key Laboratory of Cardiovascular Science, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
18
|
Yu Y, Cai Y, Yang F, Yang Y, Cui Z, Shi D, Bai R. Vascular smooth muscle cell phenotypic switching in atherosclerosis. Heliyon 2024; 10:e37727. [PMID: 39309965 PMCID: PMC11416558 DOI: 10.1016/j.heliyon.2024.e37727] [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: 05/30/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a complex pathology process involving intricate interactions among various cells and biological processes. Vascular smooth muscle cells (VSMCs) are the predominant cell type in normal arteries, and under atherosclerotic stimuli, VSMCs respond to altered blood flow and microenvironment changes by downregulating contractile markers and switching their phenotype. This review overviews the diverse phenotypes of VSMCs, including the canonical contractile VSMCs, synthetic VSMCs, and phenotypes resembling macrophages, foam cells, myofibroblasts, osteoblasts/chondrocytes, and mesenchymal stem cells. We summarize their presumed protective and pro-atherosclerotic roles in AS development. Additionally, we underscore the molecular mechanisms and regulatory pathways governing VSMC phenotypic switching, encompassing transcriptional regulation, biochemical factors, plaque microenvironment, epigenetics, miRNAs, and the cytoskeleton, emphasizing their significance in AS development. Finally, we outline probable future research directions targeting VSMCs, offering insights into potential therapeutic strategies for AS management.
Collapse
Affiliation(s)
- Yanqiao Yu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yajie Cai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Furong Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Yankai Yang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Zhuorui Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| | - Ruina Bai
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, 100091, China
| |
Collapse
|
19
|
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
|
20
|
Zhang H, Kafeiti N, Masarik K, Lee S, Yang X, Zheng H, Zhan H. Decoding Endothelial MPL and JAK2V617F Mutation: Insight Into Cardiovascular Dysfunction in Myeloproliferative Neoplasms. Arterioscler Thromb Vasc Biol 2024; 44:1960-1974. [PMID: 38989576 PMCID: PMC11335084 DOI: 10.1161/atvbaha.124.321008] [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: 03/26/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Patients with JAK2V617F-positive myeloproliferative neoplasms (MPNs) and clonal hematopoiesis of indeterminate potential face a significantly elevated risk of cardiovascular diseases. Endothelial cells carrying the JAK2V617F mutation have been detected in many patients with MPN. In this study, we investigated the molecular basis for the high incidence of cardiovascular complications in patients with MPN. METHODS We investigated the impact of endothelial JAK2V617F mutation on cardiovascular disease development using both transgenic murine models and MPN patient-derived induced pluripotent stem cell lines. RESULTS Our investigations revealed that JAK2V617F mutant endothelial cells promote cardiovascular diseases under stress, which is associated with endothelial-to-mesenchymal transition and endothelial dysfunction. Importantly, we discovered that inhibiting the endothelial TPO (thrombopoietin) receptor MPL (myeloproliferative leukemia virus oncogene) suppressed JAK2V617F-induced endothelial-to-mesenchymal transition and prevented cardiovascular dysfunction caused by mutant endothelial cells. Notably, the endothelial MPL receptor is not essential for the normal physiological regulation of blood cell counts and cardiac function. CONCLUSIONS JAK2V617F mutant endothelial cells play a critical role in the development of cardiovascular diseases in JAK2V617F-positive MPNs, and endothelial MPL could be a promising therapeutic target for preventing or ameliorating cardiovascular complications in these patients.
Collapse
Affiliation(s)
- Haotian Zhang
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
- The Graduate Program in Molecular and Cellular Biology (H. Zhang), Stony Brook University, NY
| | - Nicholas Kafeiti
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
| | - Kyla Masarik
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
| | - Sandy Lee
- Department of Molecular and Cellular Pharmacology (S.L.), Stony Brook University, NY
| | - Xiaoxi Yang
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
- Division of Rheumatology, Peking Union Medical College Hospital, Beijing, China (X.Y.)
| | - Haoyi Zheng
- Cardiac Imaging, The Heart Center, Saint Francis Hospital, Roslyn, NY (H. Zheng)
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
- Medical Service, Northport VA Medical Center, NY (H. Zhan)
| |
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
|
Dimitriadis K, Pyrpyris N, Theofilis P, Mantzouranis E, Beneki E, Kostakis P, Koutsopoulos G, Aznaouridis K, Aggeli K, Tsioufis K. Computed Tomography Angiography Identified High-Risk Coronary Plaques: From Diagnosis to Prognosis and Future Management. Diagnostics (Basel) 2024; 14:1671. [PMID: 39125547 PMCID: PMC11311283 DOI: 10.3390/diagnostics14151671] [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: 07/07/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
CT angiography has become, in recent years, a main evaluating modality for patients with coronary artery disease (CAD). Recent advancements in the field have allowed us to identity not only the presence of obstructive disease but also the characteristics of identified lesions. High-risk coronary atherosclerotic plaques are identified in CT angiographies via a number of specific characteristics and may provide prognostic and therapeutic implications, aiming to prevent future ischemic events via optimizing medical treatment or providing coronary interventions. In light of new evidence evaluating the safety and efficacy of intervening in high-risk plaques, even in non-flow-limiting disease, we aim to provide a comprehensive review of the diagnostic algorithms and implications of plaque vulnerability in CT angiography, identify any differences with invasive imaging, analyze prognostic factors and potential future therapeutic options in such patients, as well as discuss new frontiers, including intervening in non-flow-limiting stenoses and the role of CT angiography in patient stratification.
Collapse
Affiliation(s)
- Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.P.); (P.T.); (E.M.); (E.B.); (P.K.); (G.K.); (K.A.); (K.A.); (K.T.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
Chatterjee P, Martin KA. A Concept of "Athero-Oncology": Tumor-Like Smooth Muscle Cells Drive Atherosclerosis. Circulation 2024; 149:1899-1902. [PMID: 38857330 DOI: 10.1161/circulationaha.124.069446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Affiliation(s)
- Payel Chatterjee
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
25
|
Pan H, Ho SE, Xue C, Cui J, Johanson QS, Sachs N, Ross LS, Li F, Solomon RA, Connolly ES, Patel VI, Maegdefessel L, Zhang H, Reilly MP. Atherosclerosis Is a Smooth Muscle Cell-Driven Tumor-Like Disease. Circulation 2024; 149:1885-1898. [PMID: 38686559 PMCID: PMC11164647 DOI: 10.1161/circulationaha.123.067587] [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: 10/12/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Atherosclerosis, a leading cause of cardiovascular disease, involves the pathological activation of various cell types, including immunocytes (eg, macrophages and T cells), smooth muscle cells (SMCs), and endothelial cells. Accumulating evidence suggests that transition of SMCs to other cell types, known as phenotypic switching, plays a central role in atherosclerosis development and complications. However, the characteristics of SMC-derived cells and the underlying mechanisms of SMC transition in disease pathogenesis remain poorly understood. Our objective is to characterize tumor cell-like behaviors of SMC-derived cells in atherosclerosis, with the ultimate goal of developing interventions targeting SMC transition for the prevention and treatment of atherosclerosis. METHODS We used SMC lineage tracing mice and human tissues and applied a range of methods, including molecular, cellular, histological, computational, human genetics, and pharmacological approaches, to investigate the features of SMC-derived cells in atherosclerosis. RESULTS SMC-derived cells in mouse and human atherosclerosis exhibit multiple tumor cell-like characteristics, including genomic instability, evasion of senescence, hyperproliferation, resistance to cell death, invasiveness, and activation of comprehensive cancer-associated gene regulatory networks. Specific expression of the oncogenic mutant KrasG12D in SMCs accelerates phenotypic switching and exacerbates atherosclerosis. Furthermore, we provide proof of concept that niraparib, an anticancer drug targeting DNA damage repair, attenuates atherosclerosis progression and induces regression of lesions in advanced disease in mouse models. CONCLUSIONS Our findings demonstrate that atherosclerosis is an SMC-driven tumor-like disease, advancing our understanding of its pathogenesis and opening prospects for innovative precision molecular strategies aimed at preventing and treating atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Huize Pan
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sebastian E. Ho
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- These authors contributed equally
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- These authors contributed equally
| | - Jian Cui
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Quinian S. Johanson
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nadja Sachs
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- German Center for Cardiovascular Research, partner site: Munich Heart Alliance, 10785 Berlin, Germany
| | - Leila S. Ross
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Fang Li
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Robert A. Solomon
- Department of Neurologic Surgery, New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY 10032, USA
| | - E. Sander Connolly
- Department of Neurologic Surgery, New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Virendra I. Patel
- Section of Vascular Surgery and Endovascular Interventions, New York-Presbyterian Hospital/Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, 81675 Munich, Germany
- German Center for Cardiovascular Research, partner site: Munich Heart Alliance, 10785 Berlin, Germany
- Department of Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Hanrui Zhang
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Muredach P. Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
26
|
Lambert J, Oc S, Worssam MD, Häußler D, Solomon CU, Figg NL, Baxter R, Imaz M, Taylor JCK, Foote K, Finigan A, Mahbubani KT, Webb TR, Ye S, Bennett MR, Krüger A, Spivakov M, Jørgensen HF. Network-based prioritization and validation of regulators of vascular smooth muscle cell proliferation in disease. NATURE CARDIOVASCULAR RESEARCH 2024; 3:714-733. [PMID: 39215134 PMCID: PMC11182749 DOI: 10.1038/s44161-024-00474-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 04/18/2024] [Indexed: 06/21/2024]
Abstract
Aberrant vascular smooth muscle cell (VSMC) homeostasis and proliferation characterize vascular diseases causing heart attack and stroke. Here we elucidate molecular determinants governing VSMC proliferation by reconstructing gene regulatory networks from single-cell transcriptomics and epigenetic profiling. We detect widespread activation of enhancers at disease-relevant loci in proliferation-predisposed VSMCs. We compared gene regulatory network rewiring between injury-responsive and nonresponsive VSMCs, which suggested shared transcription factors but differing target loci between VSMC states. Through in silico perturbation analysis, we identified and prioritized previously unrecognized regulators of proliferation, including RUNX1 and TIMP1. Moreover, we showed that the pioneer transcription factor RUNX1 increased VSMC responsiveness and that TIMP1 feeds back to promote VSMC proliferation through CD74-mediated STAT3 signaling. Both RUNX1 and the TIMP1-CD74 axis were expressed in human VSMCs, showing low levels in normal arteries and increased expression in disease, suggesting clinical relevance and potential as vascular disease targets.
Collapse
MESH Headings
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Humans
- Cell Proliferation/genetics
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Gene Regulatory Networks
- Tissue Inhibitor of Metalloproteinase-1/metabolism
- Tissue Inhibitor of Metalloproteinase-1/genetics
- STAT3 Transcription Factor/metabolism
- STAT3 Transcription Factor/genetics
- Signal Transduction/genetics
- Cells, Cultured
- Single-Cell Analysis
- Epigenesis, Genetic
- Transcriptome
- Animals
- Core Binding Factor Alpha 2 Subunit
Collapse
Affiliation(s)
- Jordi Lambert
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Sebnem Oc
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Functional Gene Control Group, MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Matthew D Worssam
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Häußler
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, Munich, Germany
| | - Charles U Solomon
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Nichola L Figg
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Ruby Baxter
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Maria Imaz
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - James C K Taylor
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Kirsty Foote
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Alison Finigan
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Krishnaa T Mahbubani
- Collaborative Biorepository for Translational Medicine, Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Tom R Webb
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
| | - Shu Ye
- Department of Cardiovascular Sciences, University of Leicester, and National Institute for Health Research Leicester Biomedical Research Centre, Leicester, UK
- Shantou University Medical College, Shantou, China
- Cardiovascular and Metabolic Disease Translational Research Programme, National University of Singapore, Singapore, Singapore
| | - Martin R Bennett
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Achim Krüger
- TUM School of Medicine and Health, Institute of Experimental Oncology and Therapy Research, Technical University of Munich, Munich, Germany
| | - Mikhail Spivakov
- Functional Gene Control Group, MRC Laboratory of Medical Sciences, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Helle F Jørgensen
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Peng Z, Kan Q, Wang K, Deng T, Wang S, Wu R, Yao C. Deciphering smooth muscle cell heterogeneity in atherosclerotic plaques and constructing model: a multi-omics approach with focus on KLF15/IGFBP4 axis. BMC Genomics 2024; 25:490. [PMID: 38760675 PMCID: PMC11102212 DOI: 10.1186/s12864-024-10379-y] [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: 12/11/2023] [Accepted: 05/06/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Ruptured atherosclerotic plaques often precipitate severe ischemic events, such as stroke and myocardial infarction. Unraveling the intricate molecular mechanisms governing vascular smooth muscle cell (VSMC) behavior in plaque stabilization remains a formidable challenge. METHODS In this study, we leveraged single-cell and transcriptomic datasets from atherosclerotic plaques retrieved from the gene expression omnibus (GEO) database. Employing a combination of single-cell population differential analysis, weighted gene co-expression network analysis (WGCNA), and transcriptome differential analysis techniques, we identified specific genes steering the transformation of VSMCs in atherosclerotic plaques. Diagnostic models were developed and validated through gene intersection, utilizing the least absolute shrinkage and selection operator (LASSO) and random forest (RF) methods. Nomograms for plaque assessment were constructed. Tissue localization and expression validation were performed on specimens from animal models, utilizing immunofluorescence co-localization, western blot, and reverse-transcription quantitative-polymerase chain reaction (RT-qPCR). Various online databases were harnessed to predict transcription factors (TFs) and their interacting compounds, with determination of the cell-specific localization of TF expression using single-cell data. RESULTS Following rigorous quality control procedures, we obtained a total of 40,953 cells, with 6,261 representing VSMCs. The VSMC population was subsequently clustered into 5 distinct subpopulations. Analyzing inter-subpopulation cellular communication, we focused on the SMC2 and SMC5 subpopulations. Single-cell subpopulation and WGCNA analyses revealed significant module enrichments, notably in collagen-containing extracellular matrix and cell-substrate junctions. Insulin-like growth factor binding protein 4 (IGFBP4), apolipoprotein E (APOE), and cathepsin C (CTSC) were identified as potential diagnostic markers for early and advanced plaques. Notably, gene expression pattern analysis suggested that IGFBP4 might serve as a protective gene, a hypothesis validated through tissue localization and expression analysis. Finally, we predicted TFs capable of binding to IGFBP4, with Krüppel-like family 15 (KLF15) emerging as a prominent candidate showing relative specificity within smooth muscle cells. Predictions about compounds associated with affecting KLF15 expression were also made. CONCLUSION Our study established a plaque diagnostic and assessment model and analyzed the molecular interaction mechanisms of smooth muscle cells within plaques. Further analysis revealed that the transcription factor KLF15 may regulate the biological behaviors of smooth muscle cells through the KLF15/IGFBP4 axis, thereby influencing the stability of advanced plaques via modulation of the PI3K-AKT signaling pathway. This could potentially serve as a target for plaque stability assessment and therapy, thus driving advancements in the management and treatment of atherosclerotic plaques.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Gene Expression Profiling
- Gene Regulatory Networks
- Insulin-Like Growth Factor Binding Protein 4/metabolism
- Insulin-Like Growth Factor Binding Protein 4/genetics
- Kruppel-Like Transcription Factors/metabolism
- Kruppel-Like Transcription Factors/genetics
- Multiomics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/cytology
- Myocytes, Smooth Muscle/metabolism
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/pathology
- Single-Cell Analysis
- Transcriptome
- Rats
Collapse
Affiliation(s)
- Zhanli Peng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Qinghui Kan
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Kangjie Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Tang Deng
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Shenming Wang
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Ridong Wu
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
| | - Chen Yao
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, 510080, P.R. China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
| |
Collapse
|
28
|
Petrovič D, Letonja J, Petrovič D. SMAD3 rs17228212 Polymorphism Is Associated with Advanced Carotid Atherosclerosis in a Slovenian Population. Biomedicines 2024; 12:1103. [PMID: 38791063 PMCID: PMC11117620 DOI: 10.3390/biomedicines12051103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Smad proteins influence the TGFβ signaling pathway, which plays an important role in the progression of atherosclerosis. The aim of our study was to investigate the association between the rs17228212 polymorphism of the SMAD3 gene and advanced carotid atherosclerosis in Slovenian subjects and to investigate the effect of the rs17228212 SMAD3 polymorphism on the expression of SMAD3 in endarterectomy sequesters. In this cross-sectional case-control study, 881 unrelated Caucasians were divided into two groups. The first group included 308 patients with advanced carotid atherosclerosis of the common or internal carotid artery with stenosis greater than 75% that underwent a revascularization procedure (cases). The control group consisted of 573 subjects without hemodynamically significant carotid atherosclerosis. We analyzed the rs17228212 polymorphism of the SMAD3 gene using the StepOne real-time polymerase chain reaction system and TaqMan SNP genotyping assay. The results in the two genetic models showed a statistically significant association, codominant (OR 4.05; CI 1.10-17.75; p = 0.037) and dominant (OR 3.60; CI 1.15-15.45; p = 0.045). An immunohistochemical analysis of SMAD3 expression was conducted for 26 endarterectomy specimens. The T allele of the rs17228212 SMAD3 gene was shown to be associated with an increased numerical area density of SMAD3-positive cells in carotid plaques.
Collapse
Affiliation(s)
- David Petrovič
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (D.P.); (J.L.)
| | - Jernej Letonja
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (D.P.); (J.L.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Danijel Petrovič
- Laboratory for Histology and Genetics of Atherosclerosis and Microvascular Diseases, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia; (D.P.); (J.L.)
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
29
|
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
|
30
|
Mickiewicz L, Zahreddine R, Cormier K, Peries S, Del Bello A, Laffargue M, Smirnova NF. A minor tweak in transplant surgery protocols alters the cellular landscape of the arterial wall during transplant vasculopathy. FRONTIERS IN TRANSPLANTATION 2024; 3:1260125. [PMID: 38993774 PMCID: PMC11235260 DOI: 10.3389/frtra.2024.1260125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/02/2024] [Indexed: 07/13/2024]
Abstract
Introduction Transplant vasculopathy (TV) is a major complication after solid organ transplantation, distinguished by an arterial intimal thickening that obstructs the vascular lumen and leads to organ rejection. To date, TV remains largely untreatable, mainly because the processes involved in its development remain unclear. Aortic transplantation in mice, used to mimic TV, relies on highly variable experimental protocols, particularly regarding the type of anastomosis used to connect the donor aorta to the recipient. While the amount of trauma undergone by a vessel can dramatically affect the resulting pathology, the impact of the type of anastomosis on TV in mice has not been investigated in detail. Methods In this study, we compare the cellular composition of aortic grafts from BALB/C donor mice transplanted into C57BL/6J recipient mice using two different anastomosis strategies: sleeve and cuff. Results While both models recapitulated some aspects of human TV, there were striking differences in the cellular composition of the grafts. Indeed, aortic grafts from the cuff group displayed a larger coverage of the neointimal area by vascular smooth muscle cells compared to the sleeve group. Aortic grafts from the sleeve group contained higher amounts of T cells, while the cuff group displayed larger B-cell infiltrates. Discussion Together, these data indicate that a seemingly minor technical difference in transplant surgery protocols can largely impact the cellular composition of the graft, and thus the mechanisms underlying TV after aortic transplantation in mice.
Collapse
Affiliation(s)
- Laura Mickiewicz
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Rana Zahreddine
- CREFRE-Anexplo, Services Phénotypage et Microchirurgie, UMS006, INSERM, Université de Toulouse, UT3, ENVT, Toulouse, France
| | - Kévin Cormier
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Sophie Peries
- Center for Biological Ressources (Centres de Ressources Biologiques, CRB), IUCT Oncopole, Toulouse University Hospital (CHU de Toulouse), Toulouse, France
| | - Arnaud Del Bello
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
- Department of Nephrology and Organ Transplantation, Toulouse University Hospital (CHU de Toulouse), Toulouse, France
| | - Muriel Laffargue
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| | - Natalia F Smirnova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), Institut National de la Santé et de la Recherche Médicale (INSERM) U1297, University of Toulouse 3, Toulouse, France
| |
Collapse
|
31
|
Elmarasi M, Elmakaty I, Elsayed B, Elsayed A, Zein JA, Boudaka A, Eid AH. Phenotypic switching of vascular smooth muscle cells in atherosclerosis, hypertension, and aortic dissection. J Cell Physiol 2024; 239:e31200. [PMID: 38291732 DOI: 10.1002/jcp.31200] [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: 08/21/2023] [Revised: 12/12/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
Vascular smooth muscle cells (VSMCs) play a critical role in regulating vasotone, and their phenotypic plasticity is a key contributor to the pathogenesis of various vascular diseases. Two main VSMC phenotypes have been well described: contractile and synthetic. Contractile VSMCs are typically found in the tunica media of the vessel wall, and are responsible for regulating vascular tone and diameter. Synthetic VSMCs, on the other hand, are typically found in the tunica intima and adventitia, and are involved in vascular repair and remodeling. Switching between contractile and synthetic phenotypes occurs in response to various insults and stimuli, such as injury or inflammation, and this allows VSMCs to adapt to changing environmental cues and regulate vascular tone, growth, and repair. Furthermore, VSMCs can also switch to osteoblast-like and chondrocyte-like cell phenotypes, which may contribute to vascular calcification and other pathological processes like the formation of atherosclerotic plaques. This provides discusses the mechanisms that regulate VSMC phenotypic switching and its role in the development of vascular diseases. A better understanding of these processes is essential for the development of effective diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Mohamed Elmarasi
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim Elmakaty
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Basel Elsayed
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Abdelrahman Elsayed
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Jana Al Zein
- Faculty of Medical Sciences, Lebanese University, Hadath, Beirut, Lebanon
| | - Ammar Boudaka
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
32
|
Lin A, Ramaswamy Y, Misra A. Developmental heterogeneity of vascular cells: Insights into cellular plasticity in atherosclerosis? Semin Cell Dev Biol 2024; 155:3-15. [PMID: 37316416 DOI: 10.1016/j.semcdb.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/30/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
Smooth muscle cells, endothelial cells and macrophages display remarkable heterogeneity within the healthy vasculature and under pathological conditions. During development, these cells arise from numerous embryological origins, which confound with different microenvironments to generate postnatal vascular cell diversity. In the atherosclerotic plaque milieu, all these cell types exhibit astonishing plasticity, generating a variety of plaque burdening or plaque stabilizing phenotypes. And yet how developmental origin influences intraplaque cell plasticity remains largely unexplored despite evidence suggesting this may be the case. Uncovering the diversity and plasticity of vascular cells is being revolutionized by unbiased single cell whole transcriptome analysis techniques that will likely continue to pave the way for therapeutic research. Cellular plasticity is only just emerging as a target for future therapeutics, and uncovering how intraplaque plasticity differs across vascular beds may provide key insights into why different plaques behave differently and may confer different risks of subsequent cardiovascular events.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, NSW, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, NSW, Australia; Heart Research Institute, The University of Sydney, Sydney, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
33
|
Gunnersen S, Shim JT, Liu F, Tietge UJ, Sørensen CB, Bentzon JF. Conditional deletion of Ccl2 in smooth muscle cells does not reduce early atherosclerosis in mice. ATHEROSCLEROSIS PLUS 2024; 55:12-20. [PMID: 38234375 PMCID: PMC10792688 DOI: 10.1016/j.athplu.2023.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Background and aims C-C motif chemokine ligand 2 (CCL2) is a pro-inflammatory chemokine important for monocyte recruitment to the arterial wall and atherosclerotic plaques. Global knockout of Ccl2 reduces plaque formation and macrophage content in mice, but the importance of different plaque cell types in mediating this effect has not been resolved. Smooth muscle cells (SMCs) can adopt a potentially pro-inflammatory function with expression of CCL2. The present study aimed to test the hypothesis that SMC-secreted CCL2 is involved in early atherogenesis in mice. Methods SMC-restricted Cre recombinase was activated at 6 weeks of age in mice with homozygous floxed or wildtype Ccl2 alleles. Separate experiments in mice lacking the Cre recombinase transgene were conducted to control for genetic background effects. Hypercholesterolemia and atherosclerosis were induced by a tail vein injection of recombinant adeno-associated virus (rAAV) encoding proprotein convertase subtilisin/kexin type 9 (PCSK9) and a high-fat diet for 12 weeks. Results Unexpectedly, mice with SMC-specific Ccl2 deletion developed higher levels of plasma cholesterol and larger atherosclerotic plaques with more macrophages compared with wild-type littermates. When total cholesterol levels were incorporated into the statistical analysis, none of the effects on plaque development between groups remained significant. Importantly, changes in plasma cholesterol and atherosclerosis remained in mice lacking Cre recombinase indicating that they were not caused by SMC-specific CCL2 deletion but by effects of the floxed allele or passenger genes. Conclusions SMC-specific deficiency of Ccl2 does not significantly affect early plaque development in hypercholesterolemic mice.
Collapse
Affiliation(s)
- Stine Gunnersen
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Jeong Tangkjær Shim
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Fan Liu
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, 14183 Stockholm, Sweden
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands
| | - Uwe J.F. Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, 14183 Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-14186 Stockholm, Sweden
| | - Charlotte Brandt Sørensen
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Jacob Fog Bentzon
- Atherosclerosis Research Unit, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Calle de Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Palle Juul-Jensens Boulevard 11, 8200 Aarhus N, Denmark
| |
Collapse
|
34
|
Chen C, Ma J, Ren L, Sun B, Shi Y, Chen L, Wang D, Wei J, Sun Y, Cao X. Rosmarinic Acid Activates the Nrf2/ARE Signaling Pathway via the miR-25-3p/SIRT6 Axis to Inhibit Vascular Remodeling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4008-4022. [PMID: 38373191 DOI: 10.1021/acs.jafc.3c02916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
The vital pathological processes in intimal hyperplasia include aberrant vascular smooth muscle cells (VSMCs) proliferation, migration, and phenotypic switching. Rosmarinic acid (RA) is a natural phenolic acid compound. Nevertheless, the underlying mechanism of RA in neointimal hyperplasia is still unclear. Our analysis illustrated that miR-25-3p mimics significantly enhanced PDGF-BB-mediated VSMCs proliferation, migration, and phenotypic switching while RA partially weakened the effect of miR-25-3p. Mechanistically, we found that miR-25-3p directly targets sirtuin (SIRT6). The suppressive effect of the miR-25-3p inhibitor on PDGF-BB-induced VSMCs proliferation, migration, and phenotypic switch was partially eliminated by SIRT6 knockdown. The suppression of the PDGF-BB-stimulated Nrf2/ARE signaling pathway that was activated by the miR-25-3p inhibitor was exacerbated by the SIRT6 knockdown. In in vivo experiments, RA reduced the degree of intimal hyperplasia while miR-25-3p agomir partially reversed the suppressive effect of RA in vascular remodeling. Our results indicate that RA activates the Nrf2/ARE signaling pathway via the miR-25-3p/SIRT6 axis to inhibit vascular remodeling.
Collapse
Affiliation(s)
- Chen Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Jiulong Ma
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Bo Sun
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Yan Shi
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Liang Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Danqi Wang
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Jiaxin Wei
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| | - Yuan Sun
- Changsha Medical College, 1501 Leifeng Avenue, Wangcheng District, Changsha, Hunan 410000, China
| | - Xia Cao
- Department of Pharmacology, School of Pharmaceutical Sciences, Jilin University, 1266 Fujin Road, Changchun, Jilin 13002, China
| |
Collapse
|
35
|
He J, Gao Y, Yang C, Guo Y, Liu L, Lu S, He H. Navigating the landscape: Prospects and hurdles in targeting vascular smooth muscle cells for atherosclerosis diagnosis and therapy. J Control Release 2024; 366:261-281. [PMID: 38161032 DOI: 10.1016/j.jconrel.2023.12.047] [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: 09/21/2023] [Revised: 12/02/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Vascular smooth muscle cells (VSMCs) have emerged as pivotal contributors throughout all phases of atherosclerotic plaque development, effectively dispelling prior underestimations of their prevalence and significance. Recent lineage tracing studies have unveiled the clonal nature and remarkable adaptability inherent to VSMCs, thereby illuminating their intricate and multifaceted roles in the context of atherosclerosis. This comprehensive review provides an in-depth exploration of the intricate mechanisms and distinctive characteristics that define VSMCs across various physiological processes, firmly underscoring their paramount importance in shaping the course of atherosclerosis. Furthermore, this review offers a thorough examination of the significant strides made over the past two decades in advancing imaging techniques and therapeutic strategies with a precise focus on targeting VSMCs within atherosclerotic plaques, notably spotlighting meticulously engineered nanoparticles as a promising avenue. We envision the potential of VSMC-targeted nanoparticles, thoughtfully loaded with medications or combination therapies, to effectively mitigate pro-atherogenic VSMC processes. These advancements are poised to contribute significantly to the pivotal objective of modulating VSMC phenotypes and enhancing plaque stability. Moreover, our paper also delves into recent breakthroughs in VSMC-targeted imaging technologies, showcasing their remarkable precision in locating microcalcifications, dynamically monitoring plaque fibrous cap integrity, and assessing the therapeutic efficacy of medical interventions. Lastly, we conscientiously explore the opportunities and challenges inherent in this innovative approach, providing a holistic perspective on the potential of VSMC-targeted strategies in the evolving landscape of atherosclerosis research and treatment.
Collapse
Affiliation(s)
- Jianhua He
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Yu Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Can Yang
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Yujie Guo
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China
| | - Lisha Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China.
| | - Shan Lu
- School of Pharmacy, Research Center for Pharmaceutical Preparations, Hubei University of Chinese Medicine, Wuhan 430065, People's Republic of China.
| | - Hongliang He
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Sciences & Medical Engineering, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
36
|
Gong X, Liu Y, Liu H, Cao N, Zeng L, Tian M, Zeng C, Hu Y, Zhang R, Chen Y, Wu G. Re-analysis of single-cell transcriptomics reveals a critical role of macrophage-like smooth muscle cells in advanced atherosclerotic plaque. Theranostics 2024; 14:1450-1463. [PMID: 38389849 PMCID: PMC10879858 DOI: 10.7150/thno.87201] [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: 06/15/2023] [Accepted: 01/05/2024] [Indexed: 02/24/2024] Open
Abstract
Aims: Smooth muscle cell (SMC) remodeling poses a critical feature in the development and progression of atherosclerosis. Although fate mapping and in silicon approaches have expanded SMC phenotypes in atherosclerosis, it still remains elusive about the contributions of individual SMC phenotypes and molecular dynamics to advanced atherosclerotic plaque. Methods: Using single-cell transcriptome, we investigated cellular compositions of human carotid plaque laden with atherosclerotic core, followed by in vivo experiments utilizing SMC-lineage tracing technology, bulk RNA sequencing (RNA-seq) and both in vivo and in vitro validation of the underlying molecular mechanism. Results: 5 functionally distinct SMC subtypes were uncovered based on transcriptional features (described as contractile, fibroblast-like, osteogenic, synthetic and macrophage-like) within the niche. A proinflammatory, macrophage-like SMC subtype displaying an intermediary phenotype between SMC and macrophage, exhibits prominent potential in destabilizing plaque. At the molecular level, we explored cluster-specific master regulons by algorithm, and identified interferon regulatory factor-8 (IRF8) as a potential stimulator of SMC-to-macrophage transdifferentiation via activating nuclear factor-κB (NF-κB) signaling. Conclusions: Our study illustrates a comprehensive cell atlas and molecular landscape of advanced atherosclerotic lesion, which might renovate current understanding of SMC biology in atherosclerosis.
Collapse
Affiliation(s)
- Xue Gong
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- Department of Cardiology, No. 926 Hospital, Joint Logistics Support Force of PLA, P.R. China
| | - Yunchang Liu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Huiying Liu
- College of Pulmonary and Critical Care Medicine, The 8th Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Nian Cao
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Liping Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Miao Tian
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yijie Hu
- Department of Cardiac Surgery, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
| | - Runjun Zhang
- Department of Cardiology, No. 926 Hospital, Joint Logistics Support Force of PLA, P.R. China
| | - Yundai Chen
- Department of Cardiology, the Sixth Medical Centre, Chinese PLA General Hospital, Beijing, P.R. China
| | - Gengze Wu
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), P.R. China
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease Research, Ministry of Education of China; Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| |
Collapse
|
37
|
Liu H, Zhao Y, Zhao G, Deng Y, Chen YE, Zhang J. SWI/SNF Complex in Vascular Smooth Muscle Cells and Its Implications in Cardiovascular Pathologies. Cells 2024; 13:168. [PMID: 38247859 PMCID: PMC10814623 DOI: 10.3390/cells13020168] [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: 11/07/2023] [Revised: 01/09/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Mature vascular smooth muscle cells (VSMC) exhibit a remarkable degree of plasticity, a characteristic that has intrigued cardiovascular researchers for decades. Recently, it has become increasingly evident that the chromatin remodeler SWItch/Sucrose Non-Fermentable (SWI/SNF) complex plays a pivotal role in orchestrating chromatin conformation, which is critical for gene regulation. In this review, we provide a summary of research related to the involvement of the SWI/SNF complexes in VSMC and cardiovascular diseases (CVD), integrating these discoveries into the current landscape of epigenetic and transcriptional regulation in VSMC. These novel discoveries shed light on our understanding of VSMC biology and pave the way for developing innovative therapeutic strategies in CVD treatment.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
- Department of Molecular & Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yang Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| | - Guizhen Zhao
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| | - Yongjie Deng
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| | - Y. Eugene Chen
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
- Department of Cardiac Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jifeng Zhang
- Department of Internal Medicine, Cardiovascular Center, University of Michigan Medical Center, 2800 Plymouth Road, Ann Arbor, MI 48109, USA; (H.L.); (Y.Z.)
| |
Collapse
|
38
|
Lin A, Brittan M, Baker AH, Dimmeler S, Fisher EA, Sluimer JC, Misra A. Clonal Expansion in Cardiovascular Pathology. JACC Basic Transl Sci 2024; 9:120-144. [PMID: 38362345 PMCID: PMC10864919 DOI: 10.1016/j.jacbts.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 02/17/2024]
Abstract
Clonal expansion refers to the proliferation and selection of advantageous "clones" that are better suited for survival in a Darwinian manner. In recent years, we have greatly enhanced our understanding of cell clonality in the cardiovascular context. However, our knowledge of the underlying mechanisms behind this clonal selection is still severely limited. There is a transpiring pattern of clonal expansion of smooth muscle cells and endothelial cells-and, in some cases, macrophages-in numerous cardiovascular diseases irrespective of their differing microenvironments. These findings indirectly suggest the possible existence of stem-like vascular cells which are primed to respond during disease. Subsequent clones may undergo further phenotypic changes to adopt either protective or detrimental roles. By investigating these clone-forming vascular cells, we may be able to harness this inherent clonal nature for future therapeutic intervention. This review comprehensively discusses what is currently known about clonal expansion across the cardiovascular field. Comparisons of the clonal nature of vascular cells in atherosclerosis (including clonal hematopoiesis of indeterminate potential), pulmonary hypertension, aneurysm, blood vessel injury, ischemia- and tumor-induced angiogenesis, and cerebral cavernous malformations are evaluated. Finally, we discuss the potential clinical implications of these findings and propose that proper understanding and specific targeting of these clonal cells may provide unique therapeutic options for the treatment of these cardiovascular conditions.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew H. Baker
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Edward A. Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, New York, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, New York, USA
| | - Judith C. Sluimer
- Centre for Cardiovascular Science, The Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
- CARIM School for Cardiovascular Sciences, Department of Pathology, Maastricht University Medical Center (MUMC), Maastricht, the Netherlands
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
39
|
Corcoran E, Olayinka A, di Luca M, Gusti Y, Hakimjavadi R, O'Connor B, Redmond EM, Cahill PA. N-Glycans on the extracellular domain of the Notch1 receptor control Jagged-1 induced Notch signalling and myogenic differentiation of S100β resident vascular stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567576. [PMID: 38014317 PMCID: PMC10680845 DOI: 10.1101/2023.11.17.567576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Notch signalling, critical for development and postnatal homeostasis of the vascular system, is highly regulated by several mechanisms including glycosylation. While the importance of O-linked glycosylation is widely accepted, the structure and function of N-glycans has yet to be defined. Here, we take advantage of lectin binding assays in combination with pharmacological, molecular, and site-directed mutagenetic approaches to study N-glycosylation of the Notch1 receptor. We find that several key oligosaccharides containing bisecting or core fucosylated structures decorate the receptor, control expression and receptor trafficking, and dictate Jagged-1 activation of Notch target genes and myogenic differentiation of multipotent S100β vascular stem cells. N-glycans at asparagine (N) 1241 and 1587 protect the receptor from accelerated degradation, while the oligosaccharide at N888 directly affects signal transduction. Conversely, N-linked glycans at N959, N1179, N1489 do not impact canonical signalling but inhibit differentiation. Our work highlights a novel functional role for N-glycans in controlling Notch1 signalling and differentiation of vascular stem cells.
Collapse
Affiliation(s)
- Eoin Corcoran
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Abidemi Olayinka
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Mariana di Luca
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Yusof Gusti
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Roya Hakimjavadi
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Brendan O'Connor
- School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| | - Eileen M Redmond
- Department of Surgery, University of Rochester, 601 Elmwood Ave, Rochester, NY 14642, United States
| | - Paul A Cahill
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology Faculty of Science and Health, Dublin City University, Dublin, Ireland
| |
Collapse
|
40
|
Pløen GG, Sørensen CB, Bentzon JF. Severe arterial injury heals with a complex clonal structure involving a large fraction of surviving smooth muscle cells. Atherosclerosis 2023; 387:117341. [PMID: 37940399 DOI: 10.1016/j.atherosclerosis.2023.117341] [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: 02/01/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND AND AIMS Smooth muscle cell (SMC) lineage cells in atherosclerosis and flow cessation-induced neointima are oligoclonal, being recruited from a tiny fraction of medial SMCs that modulate and proliferate. The present study aimed to investigate the clonal structure of SMC lineage cells healing more severe arterial injury. METHODS Arterial injury (wire, stretch, and partial ligation) was inflicted on the right carotid artery in mice with homozygous, SMC-restricted, stochastically recombining reporter transgenes that produced mosaic expression of 10 distinguishable fluorescent phenotypes for clonal tracking. Healed arteries and contra-lateral controls were analyzed after 3 weeks. Additional analysis of cell death and proliferation after injury was performed in wildtype mice. RESULTS The total number of SMC lineage cells in healed arteries was comparable to normal arteries but comprised significantly fewer fluorescent phenotypes. The population had a complex, intermixed, clonal structure. By statistical analysis of expected versus observed fractions of fluorescent phenotypes and visual inspection of coherent groups of same-colored cells, we concluded that >98% of SMC lineage cells in healed arteries belonged to a detectable clone, indicating that nearly all surviving SMCs after severe injury at some point undergo proliferation. This was consistent with serial observations in the first week after injury, which showed severe loss of medial cells followed by widespread proliferation. CONCLUSIONS After severe arterial injury, many surviving SMCs proliferate to repair the media and form a neointima. This indicates that the fraction of medial SMCs that are mobilized to repair arteries increases with the level of injury.
Collapse
Affiliation(s)
| | | | - Jacob Fog Bentzon
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
| |
Collapse
|
41
|
Kawai K, Sakamoto A, Mokry M, Ghosh SKB, Kawakami R, Xu W, Guo L, Fuller DT, Tanaka T, Shah P, Cornelissen A, Sato Y, Mori M, Konishi T, Vozenilek AE, Dhingra R, Virmani R, Pasterkamp G, Finn AV. Clonal Proliferation Within Smooth Muscle Cells in Unstable Human Atherosclerotic Lesions. Arterioscler Thromb Vasc Biol 2023; 43:2333-2347. [PMID: 37881937 DOI: 10.1161/atvbaha.123.319479] [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: 04/20/2023] [Accepted: 10/12/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND Studies in humans and mice using the expression of an X-linked gene or lineage tracing, respectively, have suggested that clones of smooth muscle cells (SMCs) exist in human atherosclerotic lesions but are limited by either spatial resolution or translatability of the model. METHODS Phenotypic clonality can be detected by X-chromosome inactivation patterns. We investigated whether clones of SMCs exist in unstable human atheroma using RNA in situ hybridization (BaseScope) to identify a naturally occurring 24-nucleotide deletion in the 3'UTR of the X-linked BGN (biglycan) gene, a proteoglycan highly expressed by SMCs. BGN-specific BaseScope probes were designed to target the wild-type or deletion mRNA. Three different coronary artery plaque types (erosion, rupture, and adaptive intimal thickening) were selected from heterozygous females for the deletion BGN. Hybridization of target RNA-specific probes was used to visualize the spatial distribution of mutants. A clonality index was calculated from the percentage of each probe in each region of interest. Spatial transcriptomics were used to identify differentially expressed transcripts within clonal and nonclonal regions. RESULTS Less than one-half of regions of interest in the intimal plaque were considered clonal with the mean percent regions of interest with clonality higher in the intimal plaque than in the media. This was consistent for all plaque types. The relationship of the dominant clone in the intimal plaque and media showed significant concordance. In comparison with the nonclonal lesions, the regions with SMC clonality had lower expression of genes encoding cell growth suppressors such as CD74, SERF-2 (small EDRK-rich factor 2), CTSB (cathepsin B), and HLA-DPA1 (major histocompatibility complex, class II, DP alpha 1), among others. CONCLUSIONS Our novel approach to examine clonality suggests atherosclerosis is primarily a disease of polyclonally and to a lesser extent clonally expanded SMCs and may have implications for the development of antiatherosclerotic therapies.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Atsushi Sakamoto
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Michal Mokry
- Central Diagnostic Laboratory, University Medical Center Utrecht, The Netherlands (M. Mokry, G.P.)
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, Utrecht University, The Netherlands (M. Mokry)
| | - Saikat Kumar B Ghosh
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Rika Kawakami
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Weili Xu
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Liang Guo
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Daniela T Fuller
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Takamasa Tanaka
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Palak Shah
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Anne Cornelissen
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Yu Sato
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Masayuki Mori
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Takao Konishi
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Aimee E Vozenilek
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Roma Dhingra
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, University Medical Center Utrecht, The Netherlands (M. Mokry, G.P.)
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Gaithersburg, MD (K.K., A.S., S.K.B.G., R.K., W.X., L.G., D.T.F., T.T., P.S., A.C., Y.S., M. Mori, T.K., A.E.V., R.D., R.V., A.V.F.)
- University of Maryland School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
42
|
Hedin U, Chemaly M, Matic L. The more the merrier: Severe vascular injury increases engagement of smooth muscle cell clones in intimal repair. Atherosclerosis 2023; 387:117365. [PMID: 37980218 DOI: 10.1016/j.atherosclerosis.2023.117365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/20/2023]
Affiliation(s)
- Ulf Hedin
- Departments of Vascular Surgery and Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.
| | - Melody Chemaly
- Departments of Vascular Surgery and Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Ljubica Matic
- Departments of Vascular Surgery and Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
43
|
Kim SG, Sung JY, Kang YJ, Choi HC. PPARγ activation by fisetin mitigates vascular smooth muscle cell senescence via the mTORC2-FoxO3a-autophagy signaling pathway. Biochem Pharmacol 2023; 218:115892. [PMID: 37890594 DOI: 10.1016/j.bcp.2023.115892] [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: 09/12/2023] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
Cellular senescence is caused by diverse stimuli and contributes to cardiovascular diseases. Several studies have indicated that PPARγ acts as a key mediator of lipid metabolism and shown that it has a protective effect on vascular biology. Nevertheless, the mechanism responsible for the anti-aging effects of PPARγ has not been fully elucidated in vascular smooth muscle cell (VSMC). Furthermore, although mTOR complex 2 (mTORC2) is known to be involved in cellular senescence and autophagy, relatively few studies have investigated its effects as compared with mTOR complex 1 (mTORC1). Therefore, we focused on mTORC2 function and investigated the relationship between PPARγ and mTORC2, and the anti-aging mechanism in VSMC. We found PPARγ activation dose-dependently mitigated the hydrogen peroxide (H2O2)-induced senescence. Treatment of fisetin induced the translocation of PPARγ from cytosol to nuclear and inhibited VSMC senescence. Moreover, activated PPARγ increased PTEN transcription, leading to inhibition of the mTORC2 signaling pathway. We determined mTORC2 activation contributed to senescence by suppressing the FoxO3a-autophagy signaling pathway, and dual knockdown of mTORC1 and mTORC2 decreased cellular senescence and increased autophagy activation more than respective single knockdown. Finally, fisetin acted as a PPARγ activator and inhibited VSMC senescence through the mTORC2-FoxO3a-autophagy signaling pathway. These results demonstrate PPARγ is associated with cellular senescence and that fisetin has an anti-aging effect via PPARγ activation and mTORC2 inhibition in VSMC. These results demonstrate that the mTORC2 signaling pathway regulates autophagy and cellular senescence, which suggests mTORC2 should be considered a significant target for preventing cellular senescence and age-related diseases.
Collapse
Affiliation(s)
- Seul Gi Kim
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea; Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Jin Young Sung
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea; Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Young Jin Kang
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Hyoung Chul Choi
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea; Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea.
| |
Collapse
|
44
|
Ito S, Amioka N, Franklin MK, Wang P, Liang CL, Katsumata Y, Cai L, Temel RE, Daugherty A, Lu HS, Sawada H. Association of NOTCH3 With Elastic Fiber Dispersion in the Infrarenal Abdominal Aorta of Cynomolgus Monkeys. Arterioscler Thromb Vasc Biol 2023; 43:2301-2311. [PMID: 37855127 PMCID: PMC10843096 DOI: 10.1161/atvbaha.123.319244] [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: 03/05/2023] [Accepted: 10/02/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND The regional heterogeneity of vascular components and transcriptomes is an important determinant of aortic biology. This notion has been explored in multiple mouse studies. In the present study, we examined the regional heterogeneity of aortas in nonhuman primates. METHODS Aortic samples were harvested from the ascending, descending thoracic, suprarenal, and infrarenal regions of young control monkeys and adult monkeys with high fructose consumption for 3 years. The regional heterogeneity of aortic structure and transcriptomes was examined by histological and bulk RNA sequencing analyses, respectively. RESULTS Immunostaining of CD31 and αSMA (alpha-smooth muscle actin) revealed that endothelial and smooth muscle cells were distributed homogeneously across the aortic regions. In contrast, elastic fibers were less abundant and dispersed in the infrarenal aorta compared with other regions and associated with collagen deposition. Bulk RNA sequencing identified a distinct transcriptome related to the Notch signaling pathway in the infrarenal aorta with significantly increased NOTCH3 mRNA compared with other regions. Immunostaining revealed that NOTCH3 protein was increased in the media of the infrarenal aorta. The abundance of medial NOTCH3 was positively correlated with the dispersion of elastic fibers. Adult cynomolgus monkeys with high fructose consumption displayed vascular wall remodeling, such as smooth muscle cell loss and elastic fiber disruption, predominantly in the infrarenal region. The correlation between NOTCH3 and elastic fiber dispersion was enhanced in these monkeys. CONCLUSIONS Aortas of young cynomolgus monkeys display regional heterogeneity of their transcriptome and the structure of elastin and collagens. Elastic fibers in the infrarenal aorta are dispersed along with upregulation of medial NOTCH3.
Collapse
Affiliation(s)
- Sohei Ito
- Saha Cardiovascular Research Center, College of Medicine
| | - Naofumi Amioka
- Saha Cardiovascular Research Center, College of Medicine
| | | | - Pengjun Wang
- Saha Cardiovascular Research Center, College of Medicine
| | | | - Yuriko Katsumata
- Department of Biostatistics, College of Public Health, University of Kentucky, KY
- Sanders-Brown Center on Aging, University of Kentucky, KY
| | - Lei Cai
- Saha Cardiovascular Research Center, College of Medicine
| | - Ryan E. Temel
- Saha Cardiovascular Research Center, College of Medicine
- Saha Aortic Center, College of Medicine, University of Kentucky, KY
- Department of Physiology, College of Medicine, University of Kentucky, KY
| | - Alan Daugherty
- Saha Cardiovascular Research Center, College of Medicine
- Saha Aortic Center, College of Medicine, University of Kentucky, KY
- Department of Physiology, College of Medicine, University of Kentucky, KY
| | - Hong S. Lu
- Saha Cardiovascular Research Center, College of Medicine
- Saha Aortic Center, College of Medicine, University of Kentucky, KY
- Department of Physiology, College of Medicine, University of Kentucky, KY
| | - Hisashi Sawada
- Saha Cardiovascular Research Center, College of Medicine
- Saha Aortic Center, College of Medicine, University of Kentucky, KY
- Department of Physiology, College of Medicine, University of Kentucky, KY
| |
Collapse
|
45
|
Luo L, Fu C, Bell CF, Wang Y, Leeper NJ. Role of vascular smooth muscle cell clonality in atherosclerosis. Front Cardiovasc Med 2023; 10:1273596. [PMID: 38089777 PMCID: PMC10713728 DOI: 10.3389/fcvm.2023.1273596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/24/2023] [Indexed: 02/01/2024] Open
Abstract
Atherosclerotic cardiovascular disease remains the leading cause of death worldwide. While many cell types contribute to the growing atherosclerotic plaque, the vascular smooth muscle cell (SMC) is a major contributor due in part to its remarkable plasticity and ability to undergo phenotype switching in response to injury. SMCs can migrate into the fibrous cap, presumably stabilizing the plaque, or accumulate within the lesional core, possibly accelerating vascular inflammation. How SMCs expand and react to disease stimuli has been a controversial topic for many decades. While early studies relying on X-chromosome inactivation were inconclusive due to low resolution and sensitivity, recent advances in multi-color lineage tracing models have revitalized the concept that SMCs likely expand in an oligoclonal fashion during atherogenesis. Current efforts are focused on determining whether all SMCs have equal capacity for clonal expansion or if a "stem-like" progenitor cell may exist, and to understand how constituents of the clone decide which phenotype they will ultimately adopt as the disease progresses. Mechanistic studies are also beginning to dissect the processes which confer cells with their overall survival advantage, test whether these properties are attributable to intrinsic features of the expanding clone, and define the role of cross-talk between proliferating SMCs and other plaque constituents such as neighboring macrophages. In this review, we aim to summarize the historical perspectives on SMC clonality, highlight unanswered questions, and identify translational issues which may need to be considered as therapeutics directed against SMC clonality are developed as a novel approach to targeting atherosclerosis.
Collapse
Affiliation(s)
- Lingfeng Luo
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Changhao Fu
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Caitlin F. Bell
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Nicholas J. Leeper
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Cardiovascular Institute, Stanford, CA, United States
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
46
|
Belhoul-Fakir H, Brown ML, Thompson PL, Hamzah J, Jansen S. Connecting the Dots: How Injury in the Arterial Wall Contributes to Atherosclerotic Disease. Clin Ther 2023; 45:1092-1098. [PMID: 37891144 DOI: 10.1016/j.clinthera.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE The occurrence and development of atherosclerotic cardiovascular disease, which can result in severe outcomes, such as myocardial infarction, stroke, loss of limb, renal failure, and infarction of the gut, are strongly associated with injury to the intimal component of the arterial wall whether via the inside-out or outside-in pathways. The role of injury to the tunica media as a pathway of atherosclerosis initiation is an underresearched area. This review focuses on potential pathways to vessel wall injury as well as current experimental and clinical research in the middle-aged and elderly populations, including the role of exercise, as it relates to injury to the tunica media. METHODS A database search using PubMed and Google Scholar was conducted for research articles published between 1909 and 2023 that focused on pathways of atherogenesis and the impact of mechanical forces on wall injury. The following key words were searched: wall injury, tunica media, atherogenesis, vascular aging, and wall strain. Studies were analyzed, and the relevant information was extracted from each study. FINDINGS A link between high mechanical stress in the arterial wall and reduced vascular compliance was found. The stiffening and calcification of the arterial wall with aging induce high blood pressure and pulse pressure, thereby causing incident hypertension and cardiovascular disease. In turn, prolonged high mechanical stress, particularly wall strain, applied to the arterial wall during vigorous exercise, results in stiffening and calcification of tunica media, accelerated arterial aging, and cardiovascular disease events. In both scenarios, the tunica media is the primary target of mechanical stress and the first to respond to hemodynamic changes. The cyclical nature of these impacts confounds the results of each because they are not mutually exclusive. IMPLICATIONS The role of stress in the tunica media appears to be overlooked despite its relevance, and further research into new primary preventive therapies is needed aside from cautioning the role of vigorous exercise in the elderly population.
Collapse
Affiliation(s)
- Hanane Belhoul-Fakir
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia.
| | - Michael Lawrence Brown
- School of Population Health, Curtin University, Bently, Perth, Western Australia, Australia
| | - Peter L Thompson
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Juliana Hamzah
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Shirley Jansen
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Nedlands, Perth, Western Australia, Australia.
| |
Collapse
|
47
|
Hutton M, Frazer M, Lin A, Patel S, Misra A. New Targets in Atherosclerosis: Vascular Smooth Muscle Cell Plasticity and Macrophage Polarity. Clin Ther 2023; 45:1047-1054. [PMID: 37709601 DOI: 10.1016/j.clinthera.2023.08.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/07/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Despite an increase in treatment options, and substantial reductions in cardiovascular mortality over the past half-century, atherosclerosis remains the most prevalent cause of premature mortality worldwide. The development of innovative new therapies is crucial to further minimize atherosclerosis-related deaths. The diverse array of cell phenotypes derived from vascular smooth muscle cells (SMCs) and macrophages within atherosclerotic plaques are increasingly becoming recognized for their beneficial and detrimental roles in plaque stability and disease burden. This review explores how contemporary transcriptomics and fate-mapping studies have revealed vascular cell plasticity as a relatively unexplored target for therapeutic intervention. METHODS Recent literature for this narrative review was obtained by searching electronic databases (ie, Google Scholar, PubMed). Additional studies were sourced from reference lists and the authors' personal databases. FINDINGS The lipid-rich and inflammatory plaque milieu induces SMC phenotypic switching to both beneficial and detrimental phenotypes. Likewise, macrophage heterogeneity increases with disease burden to a variety of pro-inflammatory and anti-inflammatory activation states. These vascular cell phenotypes are determinants of plaque structure stability, and it is therefore highly likely that they influence clinical outcomes. Development of clinical treatments targeting deleterious phenotypes or promoting pro-healing phenotypes remains in its infancy. However, existing treatments (statins) have shown beneficial effects toward macrophage polarization, providing a rationale for more targeted approaches. In contrast, beneficial SMC phenotypic modulation with these pharmacologic agents has yet to be achieved. The range of modulated vascular cell phenotypes provides a multitude of novel targets and the potential to reduce future adverse events. IMPLICATIONS Vascular cell phenotypic heterogeneity must continue to be explored to lower cardiovascular events in the future. The rapidly increasing weight of evidence surrounding the role of SMC plasticity and macrophage polarity in plaque vulnerability provides a strong foundation upon which development of new therapeutics must follow. This approach may prove to be crucial in reducing cardiovascular events and improving patient benefit in the future.
Collapse
Affiliation(s)
- Michael Hutton
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Madeleine Frazer
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia
| | - Alexander Lin
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Royal Prince Alfred Hospital, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Ashish Misra
- Atherosclerosis and Vascular Remodeling Group, Heart Research Institute, Sydney, New South Wales, Australia; Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
48
|
Shin J, Tkachenko S, Gomez D, Tripathi R, Owens GK, Cherepanova OA. Smooth muscle cells-specific loss of OCT4 accelerates neointima formation after acute vascular injury. Front Cardiovasc Med 2023; 10:1276945. [PMID: 37942066 PMCID: PMC10627795 DOI: 10.3389/fcvm.2023.1276945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 10/09/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction There is growing evidence that smooth muscle cell (SMC) phenotypic transitions play critical roles during normal developmental and tissue recovery processes and in pathological conditions such as atherosclerosis. However, the molecular mechanisms responsible for these transitions are not well understood. Recently, we found that the embryonic stem cell/induced pluripotent stem cell (iPSC) factor OCT4, which was believed to be silenced in somatic cells, plays an atheroprotective role in SMC, and regulates angiogenesis after corneal alkali burn and hindlimb ischemia by mediating microvascular SMC and pericyte migration. However, the kinetics of OCT4 activation in arterial SMC and its role in acute pathological conditions are still unknown. Methods and Results Here, using an Oct4-IRES-GFP reporter mouse model, we found that OCT4 is reactivated in the carotid artery 18 hours post-acute ligation-induced injury, a common in vivo model of the SMC phenotypic transitions. Next, using a tamoxifen-inducible Myh11-CreERT2 Oct4 knockout mouse model, we found that the loss of OCT4, specifically in SMC, led to accelerated neointima formation and increased tunica media following carotid artery ligation, at least in part by increasing SMC proliferation within the media. Bulk RNA sequencing analysis on the cultured SMC revealed significant down-regulation of the SMC contractile markers and dysregulation of the genes belonging to the regulation of cell proliferation and, positive and negative regulation for cell migration ontological groups following genetic inactivation of Oct4. We also found that loss of Oct4 resulted in suppression of contractile SMC markers after the injury and in cultured aortic SMC. Further mechanistic studies revealed that OCT4 regulates SMC contractile genes, ACTA2 and TAGLN, at least in part by direct binding to the promoters of these genes. Conclusion These results demonstrate that the pluripotency factor OCT4 is quickly activated in SMC after the acute vascular injury and inhibits SMC hyperproliferation, which may be protective in preventing excessive neointima formation.
Collapse
Affiliation(s)
- Junchul Shin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Delphine Gomez
- Department of Medicine, Division of Cardiology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rupande Tripathi
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Olga A. Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
49
|
Revêchon G, Merino LG, Machtel P, Eriksson M. Somatic mutations in vascular wall function and age-associated disease. Eur Heart J 2023; 44:4208-4210. [PMID: 37477347 PMCID: PMC10590126 DOI: 10.1093/eurheartj/ehad454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Affiliation(s)
- Gwladys Revêchon
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7C, NEO, 14157 Huddinge, Sweden
| | - Lara Garcia Merino
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7C, NEO, 14157 Huddinge, Sweden
| | - Piotr Machtel
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7C, NEO, 14157 Huddinge, Sweden
| | - Maria Eriksson
- Department of Biosciences and Nutrition, Karolinska Institutet, Hälsovägen 7C, NEO, 14157 Huddinge, Sweden
| |
Collapse
|
50
|
Abstract
The medial layer of the arterial wall is composed mainly of vascular smooth muscle cells (VSMCs). Under physiological conditions, VSMCs assume a contractile phenotype, and their primary function is to regulate vascular tone. In contrast with terminally differentiated cells, VSMCs possess phenotypic plasticity, capable of transitioning into other cellular phenotypes in response to changes in the vascular environment. Recent research has shown that VSMC phenotypic switching participates in the pathogenesis of atherosclerosis, where the various types of dedifferentiated VSMCs accumulate in the atherosclerotic lesion and participate in the associated vascular remodeling by secreting extracellular matrix proteins and proteases. This review article discusses the 9 VSMC phenotypes that have been reported in atherosclerotic lesions and classifies them into differentiated VSMCs, intermediately dedifferentiated VSMCs, and dedifferentiated VSMCs. It also provides an overview of several methodologies that have been developed for studying VSMC phenotypic switching and discusses their respective advantages and limitations.
Collapse
Affiliation(s)
- Runji Chen
- Shantou University Medical CollegeShantouChina
| | - David G. McVey
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
| | - Daifei Shen
- Research Center for Translational MedicineThe Second Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | | | - Shu Ye
- Shantou University Medical CollegeShantouChina
- Department of Cardiovascular SciencesUniversity of LeicesterLeicesterUnited Kingdom
- Cardiovascular‐Metabolic Disease Translational Research ProgrammeNational University of SingaporeSingapore
| |
Collapse
|