801
|
Langley SR, Willeit K, Didangelos A, Matic LP, Skroblin P, Barallobre-Barreiro J, Lengquist M, Rungger G, Kapustin A, Kedenko L, Molenaar C, Lu R, Barwari T, Suna G, Yin X, Iglseder B, Paulweber B, Willeit P, Shalhoub J, Pasterkamp G, Davies AH, Monaco C, Hedin U, Shanahan CM, Willeit J, Kiechl S, Mayr M. Extracellular matrix proteomics identifies molecular signature of symptomatic carotid plaques. J Clin Invest 2017; 127:1546-1560. [PMID: 28319050 PMCID: PMC5373893 DOI: 10.1172/jci86924] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 01/19/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND. The identification of patients with high-risk atherosclerotic plaques prior to the manifestation of clinical events remains challenging. Recent findings question histology- and imaging-based definitions of the “vulnerable plaque,” necessitating an improved approach for predicting onset of symptoms. METHODS. We performed a proteomics comparison of the vascular extracellular matrix and associated molecules in human carotid endarterectomy specimens from 6 symptomatic versus 6 asymptomatic patients to identify a protein signature for high-risk atherosclerotic plaques. Proteomics data were integrated with gene expression profiling of 121 carotid endarterectomies and an analysis of protein secretion by lipid-loaded human vascular smooth muscle cells. Finally, epidemiological validation of candidate biomarkers was performed in two community-based studies. RESULTS. Proteomics and at least one of the other two approaches identified a molecular signature of plaques from symptomatic patients that comprised matrix metalloproteinase 9, chitinase 3-like-1, S100 calcium binding protein A8 (S100A8), S100A9, cathepsin B, fibronectin, and galectin-3-binding protein. Biomarker candidates measured in 685 subjects in the Bruneck study were associated with progression to advanced atherosclerosis and incidence of cardiovascular disease over a 10-year follow-up period. A 4-biomarker signature (matrix metalloproteinase 9, S100A8/S100A9, cathepsin D, and galectin-3-binding protein) improved risk prediction and was successfully replicated in an independent cohort, the SAPHIR study. CONCLUSION. The identified 4-biomarker signature may improve risk prediction and diagnostics for the management of cardiovascular disease. Further, our study highlights the strength of tissue-based proteomics for biomarker discovery. FUNDING. UK: British Heart Foundation (BHF); King’s BHF Center; and the National Institute for Health Research Biomedical Research Center based at Guy’s and St Thomas’ NHS Foundation Trust and King’s College London in partnership with King’s College Hospital. Austria: Federal Ministry for Transport, Innovation and Technology (BMVIT); Federal Ministry of Science, Research and Economy (BMWFW); Wirtschaftsagentur Wien; and Standortagentur Tirol.
Collapse
Affiliation(s)
- Sarah R. Langley
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
- Duke-NUS Medical School, Singapore
| | - Karin Willeit
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Athanasios Didangelos
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Ljubica Perisic Matic
- Department of Molecular Medicine and Surgery, Vascular Surgery, Karolinska Institute, Stockholm, Sweden
| | - Philipp Skroblin
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | | | - Mariette Lengquist
- Department of Molecular Medicine and Surgery, Vascular Surgery, Karolinska Institute, Stockholm, Sweden
| | - Gregor Rungger
- Department of Neurology, Bruneck Hospital, Bruneck, Italy
| | - Alexander Kapustin
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Ludmilla Kedenko
- First Department of Internal Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Chris Molenaar
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
- Nikon Imaging Centre, King’s College London, London, United Kingdom
| | - Ruifang Lu
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Temo Barwari
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Gonca Suna
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Xiaoke Yin
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Bernhard Iglseder
- Department of Geriatric Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Peter Willeit
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Joseph Shalhoub
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alun H. Davies
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Claudia Monaco
- Kennedy Institute, University of Oxford, Oxford, United Kingdom
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Vascular Surgery, Karolinska Institute, Stockholm, Sweden
| | - Catherine M. Shanahan
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| | - Johann Willeit
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, United Kingdom
| |
Collapse
|
802
|
Khyzha N, Alizada A, Wilson MD, Fish JE. Epigenetics of Atherosclerosis: Emerging Mechanisms and Methods. Trends Mol Med 2017; 23:332-347. [PMID: 28291707 DOI: 10.1016/j.molmed.2017.02.004] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/13/2017] [Accepted: 02/16/2017] [Indexed: 12/26/2022]
Abstract
Atherosclerosis is a vascular pathology characterized by inflammation and plaque build-up within arterial vessel walls. Vessel occlusion, often occurring after plaque rupture, can result in myocardial and cerebral infarction. Epigenetic changes are increasingly being associated with atherosclerosis and are of interest from both therapeutic and biomarker perspectives. Emerging genomic approaches that profile DNA methylation, chromatin accessibility, post-translational histone modifications, transcription factor binding, and RNA expression in low or single cell populations are poised to enhance our spatiotemporal understanding of atherogenesis. Here, we review recent therapeutically relevant epigenetic discoveries and emerging technologies that may generate new opportunities for atherosclerosis research.
Collapse
Affiliation(s)
- Nadiya Khyzha
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada
| | - Azad Alizada
- Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada; Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Michael D Wilson
- Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada; Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Canada.
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada; Heart & Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, Toronto, Canada.
| |
Collapse
|
803
|
Durgin BG, Cherepanova OA, Gomez D, Karaoli T, Alencar GF, Butcher JT, Zhou YQ, Bendeck MP, Isakson BE, Owens GK, Connelly JJ. Smooth muscle cell-specific deletion of Col15a1 unexpectedly leads to impaired development of advanced atherosclerotic lesions. Am J Physiol Heart Circ Physiol 2017; 312:H943-H958. [PMID: 28283548 PMCID: PMC5451587 DOI: 10.1152/ajpheart.00029.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 12/28/2022]
Abstract
Atherosclerotic plaque rupture with subsequent embolic events is a major cause of sudden death from myocardial infarction or stroke. Although smooth muscle cells (SMCs) produce and respond to collagens in vitro, there is no direct evidence in vivo that SMCs are a crucial source of collagens and that this impacts lesion development or fibrous cap formation. We sought to determine how conditional SMC-specific knockout of collagen type XV (COL15A1) in SMC lineage tracing mice affects advanced lesion formation given that 1) we have previously identified a Col15a1 sequence variant associated with age-related atherosclerosis, 2) COL15A1 is a matrix organizer enhancing tissue structural integrity, and 3) small interfering RNA-mediated Col15a1 knockdown increased migration and decreased proliferation of cultured human SMCs. We hypothesized that SMC-derived COL15A1 is critical in advanced lesions, specifically in fibrous cap formation. Surprisingly, we demonstrated that SMC-specific Col15a1 knockout mice fed a Western diet for 18 wk failed to form advanced lesions. SMC-specific Col15a1 knockout resulted in lesions reduced in size by 78%, with marked reductions in numbers and proliferating SMCs, and lacked a SMC and extracellular matrix-rich lesion or fibrous cap. In vivo RNA-seq analyses on SMC Col15a1 knockout and wild-type lesions suggested that a mechanism for these effects is through global repression of multiple proatherogenic inflammatory pathways involved in lesion development. These results provide the first direct evidence that a SMC-derived collagen, COL15A1, is critical during lesion pathogenesis, but, contrary to expectations, its loss resulted in marked attenuation rather than exacerbation of lesion pathogenesis.NEW & NOTEWORTHY We report the first direct in vivo evidence that a smooth muscle cell (SMC)-produced collagen, collagen type XV (COL15A1), is critical for atherosclerotic lesion development. SMC Col15a1 knockout markedly attenuated advanced lesion formation, likely through reducing SMC proliferation and impairing multiple proatherogenic inflammatory processes.
Collapse
Affiliation(s)
- Brittany G Durgin
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - Olga A Cherepanova
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - Delphine Gomez
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - Themistoclis Karaoli
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia
| | - Gabriel F Alencar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Joshua T Butcher
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - Yu-Qing Zhou
- Department of Laboratory Medicine and Pathobiology, Ted Rogers Centre for Heart Research TBEP, University of Toronto, Toronto, Ontario, Canada; and
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology, Ted Rogers Centre for Heart Research TBEP, University of Toronto, Toronto, Ontario, Canada; and
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia.,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, Virginia
| | - Jessica J Connelly
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia; .,Department of Psychology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
804
|
Ho-Tin-Noé B, Vo S, Bayles R, Ferrière S, Ladjal H, Toumi S, Deschildre C, Ollivier V, Michel JB. Cholesterol crystallization in human atherosclerosis is triggered in smooth muscle cells during the transition from fatty streak to fibroatheroma. J Pathol 2017; 241:671-682. [DOI: 10.1002/path.4873] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/01/2016] [Accepted: 12/29/2016] [Indexed: 11/12/2022]
Affiliation(s)
- Benoît Ho-Tin-Noé
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Sophie Vo
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Richard Bayles
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Stephen Ferrière
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Hayette Ladjal
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Sondes Toumi
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Catherine Deschildre
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Véronique Ollivier
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| | - Jean-Baptiste Michel
- Université Paris Diderot, Sorbonne Paris Cité; Laboratory for Vascular Translational Science; INSERM Unit 1148 Paris France
| |
Collapse
|
805
|
Roussis IM, Myers FA, Scarlett GP. RNA Whole-Mount In Situ Hybridization Proximity Ligation Assay (rISH-PLA), an Assay for Detecting RNA-Protein Complexes in Intact Cells. CURRENT PROTOCOLS IN CELL BIOLOGY 2017; 74:17.20.1-17.20.10. [PMID: 28256719 DOI: 10.1002/cpcb.13] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Techniques for studying RNA-protein interactions have lagged behind those for DNA-protein interactions as a consequence of the complexities associated with working with RNA. This unit describes a method for the adaptation of the In Situ Hybridization-Proximity Ligation Assay (ISH-PLA) to the study of RNA regulation (rISH-PLA). The rISH-PLA assay allows the identification of a given RNA-protein complex at subcellular and single-cell resolution, thus avoiding the lack of spatial resolution and sensitivity associated with assaying heterogeneous cell populations from which conventional RNA-protein interaction detection techniques suffer. This technique will be particularly usefully for studying the activity of RNA binding proteins (RBPs) in complex mixtures of cells, for example tissue sections or whole embryos. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Ioannis M Roussis
- Biophysics Laboratories, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Fiona A Myers
- Biophysics Laboratories, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Garry P Scarlett
- Biophysics Laboratories, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
806
|
Shanahan CM, Furmanik M. Endoplasmic Reticulum Stress in Arterial Smooth Muscle Cells: A Novel Regulator of Vascular Disease. Curr Cardiol Rev 2017; 13:94-105. [PMID: 27758696 PMCID: PMC5440785 DOI: 10.2174/1573403x12666161014094738] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/24/2016] [Accepted: 10/06/2016] [Indexed: 01/27/2023] Open
Abstract
Cardiovascular disease continues to be the leading cause of death in industrialised societies. The idea that the arterial smooth muscle cell (ASMC) plays a key role in regulating many vascular pathologies has been gaining importance, as has the realisation that not enough is known about the pathological cellular mechanisms regulating ASMC function in vascular remodelling. In the past decade endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) have been recognised as a stress response underlying many physiological and pathological processes in various vascular cell types. Here we summarise what is known about how ER stress signalling regulates phenotypic switching, trans/dedifferentiation and apoptosis of ASMCs and contributes to atherosclerosis, hypertension, aneurysms and vascular calcification.
Collapse
Affiliation(s)
- Catherine M Shanahan
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| | - Malgorzata Furmanik
- British Heart Foundation Centre of Research Excellence, Cardiovascular Division, James Black Centre, King's College London, 125 Coldharbour Lane, London, SE5 9NU, United Kingdom
| |
Collapse
|
807
|
Gabunia K, Herman AB, Ray M, Kelemen SE, England RN, DeLa Cadena R, Foster WJ, Elliott KJ, Eguchi S, Autieri MV. Induction of MiR133a expression by IL-19 targets LDLRAP1 and reduces oxLDL uptake in VSMC. J Mol Cell Cardiol 2017; 105:38-48. [PMID: 28257760 DOI: 10.1016/j.yjmcc.2017.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/21/2017] [Accepted: 02/27/2017] [Indexed: 10/20/2022]
Abstract
The transformation of vascular smooth muscle cells [VSMC] into foam cells leading to increased plaque size and decreased stability is a key, yet understudied step in atherogenesis. We reported that Interleukin-19 (IL-19), a novel, anti-inflammatory cytokine, attenuates atherosclerosis by anti-inflammatory effects on VSMC. In this work we report that IL-19 induces expression of miR133a, a muscle-specific miRNA, in VSMC. Although previously unreported, we report that miR133a can target and reduce mRNA abundance, mRNA stability, and protein expression of Low Density Lipoprotein Receptor Adaptor Protein 1, (LDLRAP1), an adaptor protein which functions to internalize the LDL receptor. Mutations in this gene lead to LDL receptor malfunction and cause the Autosomal Recessive Hypercholesterolemia (ARH) disorder in humans. Herein we show that IL-19 reduces lipid accumulation in VSMC, and LDLRAP1 expression and oxLDL uptake in a miR133a-dependent mechanism. We show that LDLRAP1 is expressed in plaque and neointimal VSMC of mouse and human injured arteries. Transfection of miR133a and LDLRAP1 siRNA into VSMC reduces their proliferation and uptake of oxLDL. miR133a is significantly increased in plasma from hyperlipidemic compared with normolipidemic patients. Expression of miR133a in IL-19 stimulated VSMC represents a previously unrecognized link between vascular lipid metabolism and inflammation, and may represent a therapeutic opportunity to combat vascular inflammatory diseases.
Collapse
Affiliation(s)
- Khatuna Gabunia
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Allison B Herman
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Sheri E Kelemen
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Ross N England
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Raul DeLa Cadena
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - William J Foster
- Departments of Ophthalmology & Bioengineering, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Katherine J Elliott
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Satoru Eguchi
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States.
| |
Collapse
|
808
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
809
|
Macrophages in vascular inflammation and atherosclerosis. Pflugers Arch 2017; 469:485-499. [PMID: 28168325 DOI: 10.1007/s00424-017-1941-y] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/18/2017] [Accepted: 01/23/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is characterized by lipid accumulation and chronic inflammation of the arterial wall, and its main complications-myocardial infarction and ischemic stroke-together constitute the first cause of death worldwide. Accumulation of lipid-laden macrophage foam cells in the intima of inflamed arteries has long been recognized as a hallmark of atherosclerosis. However, in recent years, an unexpected complexity in the mechanisms of macrophage accumulation in lesions, in the protective and pathogenic functions performed by macrophages and how they are regulated has been uncovered. Here, we provide an overview of the latest developments regarding the various mechanisms of macrophage accumulation in lesion, the major functional features of lesion macrophages, and how the plaque microenvironment may affect macrophage phenotype. Finally, we discuss how best to apprehend the heterogeneous ontogeny and functionality of atherosclerotic plaque macrophages and argue that moving away from a rigid nomenclature of arbitrarily defined macrophage subsets would be beneficial for research in the field.
Collapse
|
810
|
Restenosis Inhibition and Re-differentiation of TGFβ/Smad3-activated Smooth Muscle Cells by Resveratrol. Sci Rep 2017; 7:41916. [PMID: 28165488 PMCID: PMC5292946 DOI: 10.1038/srep41916] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022] Open
Abstract
To date, there is no periadventitial drug delivery method available in the clinic to prevent restenotic failure of open vascular reconstructions. Resveratrol is a promising anti-restenotic natural drug but subject to low bioavailability when systemically administered. In order to reconcile these two prominent issues, we tested effects of periadventitial delivery of resveratrol on all three major pro-restenotic pathologies including intimal hyperplasia (IH), endothelium impairment, and vessel shrinkage. In a rat carotid injury model, periadventitial delivery of resveratrol either via Pluronic gel (2-week), or polymer sheath (3-month), effectively reduced IH without causing endothelium impairment and vessel shrinkage. In an in vitro model, primary smooth muscle cells (SMCs) were stimulated with elevated transforming growth factor (TGFβ) and its signaling protein Smad3, known contributors to IH. TGFβ/Smad3 up-regulated Kruppel-like factor (KLF5) protein, and SMC de-differentiation which was reversed by KLF5 siRNA. Furthermore, TGFβ/Smad3-stimulated KLF5 production and SMC de-differentiation were blocked by resveratrol via its inhibition of the Akt-mTOR pathway. Concordantly, resveratrol attenuated Akt phosphorylation in injured arteries. Taken together, periadventitial delivery of resveratrol produces durable inhibition of all three pro-restenotic pathologies - a rare feat among existing anti-restenotic methods. Our study suggests a potential anti-restenotic modality of resveratrol application suitable for open surgery.
Collapse
|
811
|
Zhou B, Zeng S, Li N, Yu L, Yang G, Yang Y, Zhang X, Fang M, Xia J, Xu Y. Angiogenic Factor With G Patch and FHA Domains 1 Is a Novel Regulator of Vascular Injury. Arterioscler Thromb Vasc Biol 2017; 37:675-684. [PMID: 28153879 DOI: 10.1161/atvbaha.117.308992] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 01/20/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Phenotypic modulation of vascular smooth muscle cells represents a hallmark event in vascular injury. The underlying mechanism is not completely sorted out. We investigated the involvement of angiogenic factor with G patch and FHA domains 1 (Aggf1) in vascular injury focusing on the transcriptional regulation of vascular smooth muscle cell signature genes. APPROACH AND RESULTS We report here that Aggf1 expression was downregulated in several different cell models of phenotypic modulation in vitro and in the vessels after carotid artery ligation in mice. Adenovirus-mediated Aggf1 overexpression dampened vascular injury and normalized vascular smooth muscle cell signature gene expression. Mechanistically, Aggf1 interacted with myocardin and was imperative for the formation of a serum response factor-myocardin complex on gene promoters. In response to injurious stimuli, kruppel-like factor 4 was recruited to the Aggf1 promoter and enlisted histone deacetylase 11 to repress Aggf1 transcription. In accordance, depletion of kruppel-like factor 4 or histone deacetylase 11 restored Aggf1 expression and abrogated vascular smooth muscle cell phenotypic modulation. Finally, treatment of a histone deacetylase 11 inhibitor attenuated vascular injury in mice. CONCLUSIONS Therefore, we have unveiled a previously unrecognized role for Aggf1 in regulating vascular injury.
Collapse
Affiliation(s)
- Bisheng Zhou
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Sheng Zeng
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Nan Li
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Liming Yu
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Guang Yang
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Yuyu Yang
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Xinjian Zhang
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Mingming Fang
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Jun Xia
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.)
| | - Yong Xu
- From the Department of Pathophysiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, China (B.Z., S.Z., N.L., L.Y., G.Y., X.Z., Y.X.); State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing (Y.Y.); Department of Nursing, Jiangsu Jiankang Vocational University, Nanjing, China (M.F.); and Department of Respiratory Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China (J.X.).
| |
Collapse
|
812
|
Shobeiri N, Bendeck MP. Interleukin-1β Is a Key Biomarker and Mediator of Inflammatory Vascular Calcification. Arterioscler Thromb Vasc Biol 2017; 37:179-180. [DOI: 10.1161/atvbaha.116.308724] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Navid Shobeiri
- From the Department of Laboratory Medicine and Pathobiology and Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, ON, Canada
| | - Michelle P. Bendeck
- From the Department of Laboratory Medicine and Pathobiology and Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, University of Toronto, ON, Canada
| |
Collapse
|
813
|
Xia XD, Zhou Z, Yu XH, Zheng XL, Tang CK. Myocardin: A novel player in atherosclerosis. Atherosclerosis 2017; 257:266-278. [PMID: 28012646 DOI: 10.1016/j.atherosclerosis.2016.12.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 11/29/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022]
|
814
|
Shen M, Morton J, Davidge ST, Kassiri Z. Loss of smooth muscle cell disintegrin and metalloproteinase 17 transiently suppresses angiotensin II-induced hypertension and end-organ damage. J Mol Cell Cardiol 2017; 103:11-21. [DOI: 10.1016/j.yjmcc.2016.12.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/08/2016] [Accepted: 12/02/2016] [Indexed: 11/27/2022]
|
815
|
Uremia modulates the phenotype of aortic smooth muscle cells. Atherosclerosis 2017; 257:64-70. [DOI: 10.1016/j.atherosclerosis.2016.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/02/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022]
|
816
|
Brophy ML, Dong Y, Wu H, Rahman HNA, Song K, Chen H. Eating the Dead to Keep Atherosclerosis at Bay. Front Cardiovasc Med 2017; 4:2. [PMID: 28194400 PMCID: PMC5277199 DOI: 10.3389/fcvm.2017.00002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/12/2017] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is the primary cause of coronary heart disease (CHD), ischemic stroke, and peripheral arterial disease. Despite effective lipid-lowering therapies and prevention programs, atherosclerosis is still the leading cause of mortality in the United States. Moreover, the prevalence of CHD in developing countries worldwide is rapidly increasing at a rate expected to overtake those of cancer and diabetes. Prominent risk factors include the hardening of arteries and high levels of cholesterol, which lead to the initiation and progression of atherosclerosis. However, cell death and efferocytosis are critical components of both atherosclerotic plaque progression and regression, yet, few currently available therapies focus on these processes. Thus, understanding the causes of cell death within the atherosclerotic plaque, the consequences of cell death, and the mechanisms of apoptotic cell clearance may enable the development of new therapies to treat cardiovascular disease. Here, we review how endoplasmic reticulum stress and cholesterol metabolism lead to cell death and inflammation, how dying cells affect plaque progression, and how autophagy and the clearance of dead cells ameliorates the inflammatory environment of the plaque. In addition, we review current research aimed at alleviating these processes and specifically targeting therapeutics to the site of the plaque.
Collapse
Affiliation(s)
- Megan L Brophy
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Yunzhou Dong
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hao Wu
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - H N Ashiqur Rahman
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Kai Song
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hong Chen
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| |
Collapse
|
817
|
Hasanov Z, Ruckdeschel T, König C, Mogler C, Kapel SS, Korn C, Spegg C, Eichwald V, Wieland M, Appak S, Augustin HG. Endosialin Promotes Atherosclerosis Through Phenotypic Remodeling of Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2017; 37:495-505. [PMID: 28126825 DOI: 10.1161/atvbaha.116.308455] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 01/11/2017] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMC) play a key role in the pathogenesis of atherosclerosis, the globally leading cause of death. The transmembrane orphan receptor endosialin (CD248) has been characterized as an activation marker of cells of the mesenchymal lineage including tumor-associated pericytes, stromal myofibroblasts, and activated VSMC. We, therefore, hypothesized that VSMC-expressed endosialin may display functional involvement in the pathogenesis of atherosclerosis. APPROACH AND RESULTS Expression of endosialin was upregulated during atherosclerosis in apolipoprotein E (ApoE)-null mice and human atherosclerotic samples analyzed by quantitative real-time polymerase chain reaction and immunohistochemistry. Atherosclerosis, assessed by Oil Red O staining of the descending aorta, was significantly reduced in ApoE/endosialin-deficient mice on Western-type diet. Marker analysis of VSMC in lesions induced by shear stress-modifying cast implantation around the right carotid artery identified a more pronounced contractile VSMC phenotype in the absence of endosialin. Moreover, in addition to contributing to neointima formation, endosialin also potentially regulated the proinflammatory phenotype of VSMC as evidenced in surrogate cornea pocket assay experiments in vivo and corresponding flow cytometry and ELISA analyses in vitro. CONCLUSIONS The experiments identify endosialin as a potential regulator of phenotypic remodeling of VSMC contributing to atherosclerosis. The association of endosialin with atherosclerosis and its absent expression in nonatherosclerotic samples warrant further consideration of endosialin as a therapeutic target and biomarker.
Collapse
Affiliation(s)
- Zulfiyya Hasanov
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Tina Ruckdeschel
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Courtney König
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Carolin Mogler
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Stephanie S Kapel
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Claudia Korn
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Carleen Spegg
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Viktoria Eichwald
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Matthias Wieland
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Sila Appak
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.)
| | - Hellmut G Augustin
- From the Division of Vascular Oncology and Metastasis (Z.H., T.R., C.K., C.M., S.S.K., C.K., C.S., M.W., S.A., H.G.A.) and Small Animal Imaging (V.E.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany; Institute of Pathology, Technical University Munich, Germany (C.M.); Department of Vascular Biology and Tumor Angiogenesis, Medical Faculty Mannheim (CBTM), Heidelberg University, Germany (S.S.K., M.W., H.G.A.); and German Cancer Consortium, Heidelberg, Germany (H.G.A.).
| |
Collapse
|
818
|
He M, Chen Z, Martin M, Zhang J, Sangwung P, Woo B, Tremoulet AH, Shimizu C, Jain MK, Burns JC, Shyy JYJ. miR-483 Targeting of CTGF Suppresses Endothelial-to-Mesenchymal Transition: Therapeutic Implications in Kawasaki Disease. Circ Res 2017; 120:354-365. [PMID: 27923814 PMCID: PMC5391835 DOI: 10.1161/circresaha.116.310233] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/01/2016] [Accepted: 12/06/2016] [Indexed: 12/17/2022]
Abstract
RATIONALE Endothelial-mesenchymal transition (EndoMT) is implicated in myofibroblast-like cell-mediated damage to the coronary arterial wall in acute Kawasaki disease (KD) patients, as evidenced by positive staining for connective tissue growth factor (CTGF) and EndoMT markers in KD autopsy tissues. However, little is known about the molecular basis of EndoMT involved in KD. OBJECTIVE We investigated the microRNA (miRNA) regulation of CTGF and the consequent EndoMT in KD pathogenesis. As well, the modulation of this process by statin therapy was studied. METHODS AND RESULTS Sera from healthy children and KD subjects were incubated with human umbilical vein endothelial cells. Cardiovascular disease-related miRNAs, CTGF, and EndoMT markers were quantified using reverse transcriptase quantitative polymerase chain reaction, ELISA, and Western blotting. Compared with healthy controls, human umbilical vein endothelial cell incubated with sera from acute KD patients had decreased miR-483, increased CTGF, and increased EndoMT markers. Bioinformatics analysis followed by functional validation demonstrated that Krüppel-like factor 4 (KLF4) transactivates miR-483, which in turn targets the 3' untranslated region of CTGF mRNA. Overexpression of KLF4 or pre-miR-483 suppressed, whereas knockdown of KLF4 or anti-miR-483 enhanced, CTGF expression in endothelial cells in vitro and in vivo. Furthermore, atorvastatin, currently being tested in a phase I/IIa clinical trial in KD children, induced KLF4-miR-483, which suppressed CTGF and EndoMT in endothelial cells. CONCLUSIONS KD sera suppress the KLF4-miR-483 axis in endothelial cells, leading to increased expression of CTGF and induction of EndoMT. This detrimental process in the endothelium may contribute to coronary artery abnormalities in KD patients. Statin therapy may benefit acute KD patients, in part, through the restoration of KLF4-miR-483 expression. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifier: NCT01431105.
Collapse
Affiliation(s)
- Ming He
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Zhen Chen
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Marcy Martin
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Jin Zhang
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Panjamaporn Sangwung
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Brian Woo
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Adriana H Tremoulet
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Chisato Shimizu
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Mukesh K Jain
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.)
| | - Jane C Burns
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.).
| | - John Y-J Shyy
- From the Cardiovascular Research Center, Key Laboratory of Environment and Genes Related to Diseases (M.H., J.Z., J.Y.-J.S.), Department of Rheumatology, First Affiliated Hospital (M.H.), Xi'an Jiaotong University Health Science Center, China; Division of Cardiology, Department of Medicine (M.H., Z.C., M.M., B.W., J.Y.-J.S.) and Department of Pediatrics (A.H.T., C.S., J.C.B.), University of California, San Diego; Rady Children's Hospital, San Diego, CA (A.H.T., J.C.B.); Division of Biochemistry and Molecular Biology, University of California, Riverside (M.M.); and Case Cardiovascular Research Institute, School of Medicine, Case Western Reserve University, Cardiovascular Research Center, Cleveland, OH (P.S., M.K.J.).
| |
Collapse
|
819
|
Fujishima Y, Maeda N, Matsuda K, Masuda S, Mori T, Fukuda S, Sekimoto R, Yamaoka M, Obata Y, Kita S, Nishizawa H, Funahashi T, Ranscht B, Shimomura I. Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis. FASEB J 2017; 31:1571-1583. [PMID: 28062540 DOI: 10.1096/fj.201601064r] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/19/2016] [Indexed: 12/20/2022]
Abstract
Adiponectin, an adipocyte-derived protein abundant in the circulation, is thought to be protective against atherosclerosis. However, it is not fully understood how the association of adiponectin with vascular cells and its antiatherogenic effect are connected. In this study, T-cadherin was essential for accumulation of adiponectin in the neointima and atherosclerotic plaque lesions, and the adiponectin-T-cadherin association protected against vascular injury. In the apolipoprotein E-knockout (ApoE-KO) mice, adiponectin and T-cadherin colocalized on endothelial cells and synthetic smooth muscle cells in the aortic intima. Notably, aortic adiponectin protein disappeared in T-cadherin/ApoE double-knockout (Tcad/ApoE-DKO) mice with significant elevation of blood adiponectin concentration. Furthermore, in Tcad/ApoE-DKO mice, carotid artery ligation resulted in a significant increase of neointimal thickness compared with ApoE-KO mice. Finally, on a high-cholesterol diet, Tcad/ApoE-DKO mice increased atherosclerotic plaque formation, despite a 5-fold increase in plasma adiponectin level compared with that in ApoE-KO mice. In vitro, knockdown of T-cadherin from human aortic smooth muscle cells (HASMCs) with synthetic phenotype significantly reduced adiponectin accumulation on HASMCs and negated the inhibitory effect of adiponectin on proinflammatory change. Collective evidence showed that adiponectin accumulates in the vasculature via T-cadherin, and the adiponectin-T-cadherin association plays a protective role against neointimal and atherosclerotic plaque formations.-Fujishima, Y., Maeda, N., Matsuda, K., Masuda, S., Mori, T., Fukuda, S., Sekimoto, R., Yamaoka, M., Obata, Y., Kita, S., Nishizawa, H., Funahashi, T., Ranscht, B., Shimomura, I. Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis.
Collapse
Affiliation(s)
- Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan; .,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan; and
| | - Keisuke Matsuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shigeki Masuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takuya Mori
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shiro Fukuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryohei Sekimoto
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Masaya Yamaoka
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshinari Obata
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shunbun Kita
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan; and
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tohru Funahashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan; and
| | - Barbara Ranscht
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
820
|
Marzolla V, Armani A, Mammi C, Moss ME, Pagliarini V, Pontecorvo L, Antelmi A, Fabbri A, Rosano G, Jaffe IZ, Caprio M. Essential role of ICAM-1 in aldosterone-induced atherosclerosis. Int J Cardiol 2017; 232:233-242. [PMID: 28089144 DOI: 10.1016/j.ijcard.2017.01.013] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/25/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Elevated aldosterone is associated with increased risk of atherosclerosis complications, whereas treatment with mineralocorticoid receptor (MR) antagonists decreases the rate of cardiovascular events. Here we test the hypothesis that aldosterone promotes early atherosclerosis by modulating intercellular adhesion molecule-1 (ICAM-1) expression and investigate the molecular mechanisms by which aldosterone regulates ICAM-1 expression. METHODS AND RESULTS Apolipoprotein-E (ApoE)-/- mice fed an atherogenic diet and treated with aldosterone for 4weeks showed increased vascular expression of ICAM-1, paralleled by enhanced atherosclerotic plaque size in the aortic root. Moreover, aldosterone treatment resulted in increased plaque lipid and inflammatory cell content, consistent with an unstable plaque phenotype. ApoE/ICAM-1 double knockout (ApoE-/-/ICAM-1-/-) littermates were protected from the aldosterone-induced increase in plaque size, lipid content and macrophage infiltration. Since aldosterone is known to regulate ICAM-1 transcription via MR in human endothelial cells, we explored MR regulation of the ICAM-1 promoter. Luciferase reporter assays performed in HUVECs using deletion constructs of the human ICAM-1 gene promoter showed that a region containing a predicted MR-responsive element (MRE) is required for MR-dependent transcriptional regulation of ICAM-1. CONCLUSIONS Pro-atherogenic effects of aldosterone are mediated by increased ICAM-1 expression, through transcriptional regulation by endothelial MR. These data enhance our understanding of the molecular mechanism by which MR activation promotes atherosclerosis complications.
Collapse
Affiliation(s)
- Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Mary E Moss
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Vittoria Pagliarini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy; Laboratory of Neuroembryology, Fondazione Santa Lucia, 00143 Rome, Italy
| | - Laura Pontecorvo
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Antonella Antelmi
- Interinstitutional Multidisciplinary Biobank (BioBIM), IRCCS San Raffaele Pisana, 00166 Rome, Italy
| | - Andrea Fabbri
- Department of Systems Medicine, Endocrinology Unit, S. Eugenio & CTO A. Alesini Hospitals-ASL RM2, University Tor Vergata, Rome, Italy
| | - Giuseppe Rosano
- Cardiovascular & Cell Science Institute, St George's Hospital NHS Trust, University of London, London, United Kingdom; Department of Medical Sciences, IRCCS San Raffaele, Rome, Italy
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, USA
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
821
|
Liberman M, Marti LC. Vascular Calcification Regulation by Exosomes in the Vascular Wall. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 998:151-160. [DOI: 10.1007/978-981-10-4397-0_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
822
|
Gonzalez L, Trigatti BL. Macrophage Apoptosis and Necrotic Core Development in Atherosclerosis: A Rapidly Advancing Field with Clinical Relevance to Imaging and Therapy. Can J Cardiol 2016; 33:303-312. [PMID: 28232016 DOI: 10.1016/j.cjca.2016.12.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/14/2016] [Accepted: 12/14/2016] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular diseases represent 1 of the main causes of death worldwide, and atherosclerosis is 1 of the major contributors leading to ischemic heart disease. Macrophages actively participate in all stages of atherosclerosis development, from plaque initiation to the transition to vulnerable plaques. Macrophage apoptosis, in particular, has been recognized as a critical step in the formation of the necrotic core, a key characteristic of unstable lesions. In this review, we discuss the role of macrophage apoptosis and clearance of apoptotic cells by efferocytosis in the development of atherosclerosis, with particular emphasis on their contribution to the development of the necrotic core and the clinical implications of this process for plaque stabilization. We consider the molecular triggers of macrophage apoptosis during atherogenesis, the role of endoplasmic reticulum (ER) stress, the roles of key cellular mediators of apoptosis and efferocytosis, and mechanisms of defective efferocytosis in the progression of atherosclerotic plaques. Finally, we discuss the important clinical implications of rapidly evolving macrophage science, such as novel approaches to imaging vulnerable atherosclerotic plaques with macrophage-sensitive positron emission tomography and magnetic resonance imaging, the role of macrophages in mediating beneficial pleiotropic actions of lipid-lowering therapies, and novel therapeutic modalities targeting ER stress, autophagy, and deficient efferocytosis. Advances in understanding the critical role of macrophages in the progression and destabilization of atherosclerosis have the potential to greatly improve the prevention and management of atherosclerotic diseases over the next decade.
Collapse
Affiliation(s)
- Leticia Gonzalez
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Bernardo Louis Trigatti
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
823
|
Bisgaard LS, Bosteen MH, Fink LN, Sørensen CM, Rosendahl A, Mogensen CK, Rasmussen SE, Rolin B, Nielsen LB, Pedersen TX. Liraglutide Reduces Both Atherosclerosis and Kidney Inflammation in Moderately Uremic LDLr-/- Mice. PLoS One 2016; 11:e0168396. [PMID: 27992511 PMCID: PMC5161477 DOI: 10.1371/journal.pone.0168396] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 11/29/2016] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) leads to uremia. CKD is characterized by a gradual increase in kidney fibrosis and loss of kidney function, which is associated with a progressive increase in risk of atherosclerosis and cardiovascular death. To prevent progression of both kidney fibrosis and atherosclerosis in uremic settings, insight into new treatment options with effects on both parameters is warranted. The GLP-1 analogue liraglutide improves glucose homeostasis, and is approved for treatment of type 2 diabetes. Animal studies suggest that GLP-1 also dampens inflammation and atherosclerosis. Our aim was to examine effects of liraglutide on kidney fibrosis and atherosclerosis in a mouse model of moderate uremia (5/6 nephrectomy (NX)). Uremic (n = 29) and sham-operated (n = 14) atherosclerosis-prone low density lipoprotein receptor knockout mice were treated with liraglutide (1000 μg/kg, s.c. once daily) or vehicle for 13 weeks. As expected, uremia increased aortic atherosclerosis. In the remnant kidneys from NX mice, flow cytometry revealed an increase in the number of monocyte-like cells (CD68+F4/80-), CD4+, and CD8+ T-cells, suggesting that moderate uremia induced kidney inflammation. Furthermore, markers of fibrosis (i.e. Col1a1 and Col3a1) were upregulated, and histological examinations showed increased glomerular diameter in NX mice. Importantly, liraglutide treatment attenuated atherosclerosis (~40%, p < 0.05) and reduced kidney inflammation in NX mice. There was no effect of liraglutide on expression of fibrosis markers and/or kidney histology. This study suggests that liraglutide has beneficial effects in a mouse model of moderate uremia by reducing atherosclerosis and attenuating kidney inflammation.
Collapse
Affiliation(s)
- Line S. Bisgaard
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Global Research, Novo Nordisk, Måløv, Denmark
| | - Markus H. Bosteen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | | | | | | | - Bidda Rolin
- Global Research, Novo Nordisk, Måløv, Denmark
| | - Lars B. Nielsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Tanja X. Pedersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
824
|
Ruparelia N, Chai JT, Fisher EA, Choudhury RP. Inflammatory processes in cardiovascular disease: a route to targeted therapies. Nat Rev Cardiol 2016; 14:133-144. [PMID: 27905474 DOI: 10.1038/nrcardio.2016.185] [Citation(s) in RCA: 343] [Impact Index Per Article: 38.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammatory processes are firmly established as central to the development and complications of cardiovascular diseases. Elevated levels of inflammatory markers have been shown to be predictive of future cardiovascular events. The specific targeting of these processes in experimental models has been shown to attenuate myocardial and arterial injury, reduce disease progression, and promote healing. However, the translation of these observations and the demonstration of clear efficacy in clinical practice have been disappointing. A major limitation might be that tools currently used to measure 'inflammation' are insufficiently precise and do not provide information about disease site and activity, or discriminate between functionally important activation pathways. The challenge, therefore, is to make measures of inflammation that are more meaningful, and which can guide specific targeted therapies. In this Review, we consider the roles of inflammatory processes in the related pathologies of atherosclerosis and acute myocardial infarction, by providing an evaluation of the known and emerging inflammatory pathways. We highlight contemporary techniques to characterize and quantify inflammation, and consider how they might be used to guide specific treatments. Finally, we discuss emerging opportunities in the field, including their current limitations and challenges that are the focus of ongoing study.
Collapse
Affiliation(s)
- Neil Ruparelia
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK
| | - Joshua T Chai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,Acute Vascular Imaging Centre, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Edward A Fisher
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,The Center for the Prevention of Cardiovascular Disease and the Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York 10016, USA
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,Acute Vascular Imaging Centre, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| |
Collapse
|
825
|
Wang K, Li W, Yu Q, Guo B, Yang B, Zhang C, Li M, Li J, Hu S, Zheng Q, Song Z. High Mobility Group Box 1 Mediates Interferon-γ-Induced Phenotypic Modulation of Vascular Smooth Muscle Cells. J Cell Biochem 2016; 118:518-529. [PMID: 27579780 DOI: 10.1002/jcb.25682] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 08/26/2016] [Indexed: 02/06/2023]
Abstract
The phenotypic modulation of VSMCs is a key cellular event driving neointimal formation and vascular remodeling. As a multifaceted cytokine of cell-mediated immunity, IFN-γ has been shown to play a critical role in the pathogenesis of vascular proliferative diseases. Although the important function of IFN-γ on regulating VSMC activation is well established, the molecular mechanisms by which elicits VSMC responses are poorly defined. Recent studies have identified HMGB1 as a principal effector to mediate IFN-γ-dependent biological functions in multiple cell types. Moreover, SIRT1 has emerged as a critical regulator of cellular processes through deacetylating multiple substrates, including HMGB1. Thus, we examined the role of IFN-γ on HMGB1 release, SIRT1 expression, and VSMC phenotypic modulation as well as the underlying molecular mechanisms. We show that IFN-γ dose-dependently induces HMGB1 cytoplasmic accumulation and its active release from VSMCs, resulting in enhanced HMGB1 in the medium. Conversely, IFN-γ treatment led to a dramatic decrease in SIRT1 expression. Additionally, pretreatment with resveratrol, a selective SIRT1 activator, abrogated IFN-γ-induced HMGB1 translocation and its release. Moreover, IFN-γ stimulates VSMC phenotypic modulation to an activated synthetic state characterized by the repression of SMC differentiation markers such as SM22α and calponin and the increase in cell motility. In contrast, blocking HMGB1 release or activity by resveratrol and HMGB1-neutralizing antibody prevents IFN-γ-induced phenotypic modulation of VSMCs. Overall, this study provides the first evidence showing that HMGB1 plays a critical role in regulating VSMC phenotypic modulation, suggesting that HMGB1 may be a potential therapeutic target to prevent vascular occlusive diseases. J. Cell. Biochem. 118: 518-529, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kun Wang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Li
- Department of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qihong Yu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bing Guo
- Aab Cardiovascular Research Institute, University of Rochester, School of Medicine and Dentistry, Rochester, New York
| | - Bin Yang
- Second Department of Hepatobiliary Surgery, Zhujiang Hospital, State Key Laboratory of Organ Failure Research, Co-Innovation Center for Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Chen Zhang
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Li
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaobo Hu
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qichang Zheng
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zifang Song
- Department of Hepatobiliary Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
826
|
Zernecke A. CD98 promotes vascular smooth muscle cell accumulation in atherosclerosis to confer plaque stability. Atherosclerosis 2016; 256:128-130. [PMID: 27939649 DOI: 10.1016/j.atherosclerosis.2016.11.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/24/2016] [Indexed: 11/19/2022]
Affiliation(s)
- Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Josef-Schneider-Str. 2, D16, 97080 Würzburg, Germany.
| |
Collapse
|
827
|
Sunaga H, Matsui H, Anjo S, Syamsunarno MRAA, Koitabashi N, Iso T, Matsuzaka T, Shimano H, Yokoyama T, Kurabayashi M. Elongation of Long-Chain Fatty Acid Family Member 6 (Elovl6)-Driven Fatty Acid Metabolism Regulates Vascular Smooth Muscle Cell Phenotype Through AMP-Activated Protein Kinase/Krüppel-Like Factor 4 (AMPK/KLF4) Signaling. J Am Heart Assoc 2016; 5:e004014. [PMID: 27881420 PMCID: PMC5210431 DOI: 10.1161/jaha.116.004014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 10/17/2016] [Indexed: 01/16/2023]
Abstract
BACKGROUND Fatty acids constitute the critical components of cell structure and function, and dysregulation of fatty acid composition may exert diverging vascular effects including proliferation, migration, and differentiation of vascular smooth muscle cells (VSMCs). However, direct evidence for this hypothesis has been lacking. We investigated the role of elongation of long-chain fatty acid member 6 (Elovl6), a rate-limiting enzyme catalyzing the elongation of saturated and monounsaturated long-chain fatty acid, in the regulation of phenotypic switching of VSMC. METHODS AND RESULTS Neointima formation following wire injury was markedly inhibited in Elovl6-null (Elovl6-/-) mice, and cultured VSMCs with siRNA-mediated knockdown of Elovl6 was barely responsive to PDGF-BB. Elovl6 inhibition induced cell cycle suppressors p53 and p21 and reduced the mammalian targets of rapamycin (mTOR) phosphorylation and VSMC marker expression. These changes are ascribed to increased palmitate levels and reduced oleate levels, changes that lead to reactive oxygen species (ROS) production and resulting AMP-activated protein kinase (AMPK) activation. Notably, Elovl6 inhibition robustly induced the pluripotency gene Krüppel-like factor 4 (KLF4) expression in VSMC, and KLF4 knockdown significantly attenuated AMPK-induced phenotypic switching of VSMC, indicating that KLF4 is a bona fide target of AMPK. CONCLUSIONS We demonstrate for the first time that dysregulation of Elovl6-driven long-chain fatty acid metabolism induces phenotypic switching of VSMC via ROS production and AMPK/KLF4 signaling that leads to growth arrest and downregulation of VSMC marker expression. The modulation of Elovl6-mediated cellular processes may provide an intriguing approach for tackling atherosclerosis and postangioplasty restenosis.
Collapse
Affiliation(s)
- Hiroaki Sunaga
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
- Department of Medicine and Biological Sciences, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroki Matsui
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Saki Anjo
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Mas Risky A A Syamsunarno
- Department of Medicine and Biological Sciences, Gunma University Graduate School of Medicine, Maebashi, Japan
- Department of Biochemistry, Faculty of Medicine Universitas Padjadjaran, Jatinangor, Indonesia
| | - Norimichi Koitabashi
- Department of Medicine and Biological Sciences, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tatsuya Iso
- Department of Medicine and Biological Sciences, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Takashi Matsuzaka
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Graduate School of Comprehensive Human Sciences International Institute for Integrative Sleep Medicine (WPI-IIIS), Tsukuba, Japan
| | - Tomoyuki Yokoyama
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Maebashi, Japan
| | - Masahiko Kurabayashi
- Department of Medicine and Biological Sciences, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
828
|
Richards J, Ogoe HA, Li W, Babayewa O, Xu W, Bythwood T, Garcia-Barrios M, Ma L, Song Q. DNA Methylation Signature of Post-injury Neointimal Cells During Vascular Remodeling in the Rat Balloon Injury Model. ACTA ACUST UNITED AC 2016; 5. [PMID: 27857867 PMCID: PMC5110257 DOI: 10.4172/2168-9547.1000163] [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] [Indexed: 11/13/2022]
Abstract
Vascular smooth muscle cell (VSMC) accumulation in the neointimal is a common feature in vascular diseases such as atherosclerosis, transplant arteriosclerosis and restenosis. In this study, we isolated the neointimal cells and uninjured residential vascular smooth muscle cells by laser micro dissection and carried out single-cell whole-genome methylation sequencing. We also sequenced the bisulfite converted genome of circulating bone-marrow-derived cells such as peripheral blood mononuclear cells (PBMC) and bone marrow mononuclear cells (BMMC). We found totally 2,360 differential methylation sites (DMS) annotated to 1,127 gene regions. The majority of differentially methylated regions (DMRs) were located in intergenic regions, outside those CpG islands and island shores. Interestingly, exons have less DMRs than promotors and introns, and CpG islands contain more DMRs than islands shores. Pearson correlation analysis showed a clear clustering of neointimal cells with PBMC/BMMC. Gene set enrichment analysis of differentially methylated CpG sites revealed that many genes were important for regulation of VSMC differentiation and stem cell maintenance. In conclusion, our results showed that neointimal cells are more similar to the progenitor cells in methylation profile than the residential VSMCs at the 30th day after the vascular injury.
Collapse
Affiliation(s)
- Jendai Richards
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Henry Ato Ogoe
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Wenzhi Li
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Oguljahan Babayewa
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Wei Xu
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Tameka Bythwood
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Minerva Garcia-Barrios
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Li Ma
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA; 4DGenome Inc, Atlanta, Georgia, USA
| | - Qing Song
- Cardiovascular Research Institute and Department of Medicine, Morehouse School of Medicine, Atlanta, Georgia, USA; 4DGenome Inc, Atlanta, Georgia, USA
| |
Collapse
|
829
|
Majesky MW, Horita H, Ostriker A, Lu S, Regan JN, Bagchi A, Dong XR, Poczobutt J, Nemenoff RA, Weiser-Evans MCM. Differentiated Smooth Muscle Cells Generate a Subpopulation of Resident Vascular Progenitor Cells in the Adventitia Regulated by Klf4. Circ Res 2016; 120:296-311. [PMID: 27834190 DOI: 10.1161/circresaha.116.309322] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/31/2016] [Accepted: 11/09/2016] [Indexed: 01/31/2023]
Abstract
RATIONALE The vascular adventitia is a complex layer of the vessel wall consisting of vasa vasorum microvessels, nerves, fibroblasts, immune cells, and resident progenitor cells. Adventitial progenitors express the stem cell markers, Sca1 and CD34 (adventitial sca1-positive progenitor cells [AdvSca1]), have the potential to differentiate in vitro into multiple lineages, and potentially contribute to intimal lesions in vivo. OBJECTIVE Although emerging data support the existence of AdvSca1 cells, the goal of this study was to determine their origin, degree of multipotency and heterogeneity, and contribution to vessel remodeling. METHODS AND RESULTS Using 2 in vivo fate-mapping approaches combined with a smooth muscle cell (SMC) epigenetic lineage mark, we report that a subpopulation of AdvSca1 cells is generated in situ from differentiated SMCs. Our data establish that the vascular adventitia contains phenotypically distinct subpopulations of progenitor cells expressing SMC, myeloid, and hematopoietic progenitor-like properties and that differentiated SMCs are a source to varying degrees of each subpopulation. SMC-derived AdvSca1 cells exhibit a multipotent phenotype capable of differentiating in vivo into mature SMCs, resident macrophages, and endothelial-like cells. After vascular injury, SMC-derived AdvSca1 cells expand in number and are major contributors to adventitial remodeling. Induction of the transcription factor Klf4 in differentiated SMCs is essential for SMC reprogramming in vivo, whereas in vitro approaches demonstrate that Klf4 is essential for the maintenance of the AdvSca1 progenitor phenotype. CONCLUSIONS We propose that generation of resident vascular progenitor cells from differentiated SMCs is a normal physiological process that contributes to the vascular stem cell pool and plays important roles in arterial homeostasis and disease.
Collapse
Affiliation(s)
- Mark W Majesky
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.).
| | - Henrick Horita
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Allison Ostriker
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Sizhao Lu
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Jenna N Regan
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Ashim Bagchi
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Xiu Rong Dong
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Joanna Poczobutt
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Raphael A Nemenoff
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.)
| | - Mary C M Weiser-Evans
- From the Division of Renal Diseases and Hypertension (H.H., A.O., S.L., A.B., J.P., R.A.N., M.C.M.W.-E.) and Cardiovascular Pulmonary Research Program, Division of Cardiology (R.A.N., M.C.M.W.-E.), School of Medicine, Consortium for Fibrosis Research and Translation, University of Colorado, Aurora (M.C.M.W.-E.); Division of Endocrinology, Department of Medicine, Indiana University, Indianapolis (J.N.R.); Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, WA (M.W.M., X.R.D.); and Department of Pediatrics and Department of Pathology, University of Washington, Seattle (M.W.M.).
| |
Collapse
|
830
|
Cao Y, Long J, Ji Y, Chen G, Shen Y, Gong Y, Li J. Foam cell formation by particulate matter (PM) exposure: a review. Inhal Toxicol 2016; 28:583-590. [PMID: 27706953 DOI: 10.1080/08958378.2016.1236157] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Increasing evidence suggests that exposure of particulate matter (PM) from traffic vehicles, e.g., diesel exhaust particles (DEP), was associated with adverse vascular effects, e.g., acceleration of atherosclerotic plaque progression. By analogy, engineered nanoparticles (NPs) could also induce similar effects. The formation of lipid laden foam cells, derived predominately from macrophages and vascular smooth muscle cells (VSMC), is closely associated with the development of atherosclerosis and adverse vascular effects. We reviewed current studies about particle exposure-induced lipid laden foam cell formation. In vivo studies using animal models have shown that exposure of air pollution by PM promoted lipid accumulation in alveolar macrophages or foam cells in plaques, which was likely associated with pulmonary inflammation or systemic oxidative stress, but not blood lipid profile. In support of these findings, in vitro studies showed that direct exposure of cultured macrophages to DEP or NP exposure, with or without further exposure to external lipids, promoted intracellular lipid accumulation. The mechanisms remained unknown. Although a number studies found increased reactive oxygen species (ROS) or an adaptive response to oxidative stress, the exact role of oxidative stress in mediating particle-induced foam cell formation requires future research. There is currently lack of reports concerning VSMC as a source for foam cells induced by particle exposure. In the future, it is necessary to explore the role of foam cell formation in particle exposure-induced atherosclerosis development. In addition, the formation of VSMC derived foam cells by particle exposure may also need extensive studies.
Collapse
Affiliation(s)
- Yi Cao
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| | - Jimin Long
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| | - Yuejia Ji
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| | - Gui Chen
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| | - Yuexin Shen
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| | - Yu Gong
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| | - Juan Li
- a Key Laboratory of Environment-Friendly Chemistry and Applications of Ministry Education, Laboratory of Biochemistry, College of Chemistry, Xiangtan University , Xiangtan , China
| |
Collapse
|
831
|
Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen JM. Senescent intimal foam cells are deleterious at all stages of atherosclerosis. Science 2016; 354:472-477. [PMID: 27789842 DOI: 10.1126/science.aaf6659] [Citation(s) in RCA: 834] [Impact Index Per Article: 92.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 09/29/2016] [Indexed: 12/11/2022]
Abstract
Advanced atherosclerotic lesions contain senescent cells, but the role of these cells in atherogenesis remains unclear. Using transgenic and pharmacological approaches to eliminate senescent cells in atherosclerosis-prone low-density lipoprotein receptor-deficient (Ldlr-/-) mice, we show that these cells are detrimental throughout disease pathogenesis. We find that foamy macrophages with senescence markers accumulate in the subendothelial space at the onset of atherosclerosis, where they drive pathology by increasing expression of key atherogenic and inflammatory cytokines and chemokines. In advanced lesions, senescent cells promote features of plaque instability, including elastic fiber degradation and fibrous cap thinning, by heightening metalloprotease production. Together, these results demonstrate that senescent cells are key drivers of atheroma formation and maturation and suggest that selective clearance of these cells by senolytic agents holds promise for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Bennett G Childs
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Darren J Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Tobias Wijshake
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA. Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, Netherlands
| | - Cheryl A Conover
- Division of Endocrinology, Metabolism, and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA. Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
832
|
Mazurek R, Dave JM, Chandran RR, Misra A, Sheikh AQ, Greif DM. Vascular Cells in Blood Vessel Wall Development and Disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:323-350. [PMID: 28212800 DOI: 10.1016/bs.apha.2016.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The vessel wall is composed of distinct cellular layers, yet communication among individual cells within and between layers results in a dynamic and versatile structure. The morphogenesis of the normal vascular wall involves a highly regulated process of cell proliferation, migration, and differentiation. The use of modern developmental biological and genetic approaches has markedly enriched our understanding of the molecular and cellular mechanisms underlying these developmental events. Additionally, the application of similar approaches to study diverse vascular diseases has resulted in paradigm-shifting insights into pathogenesis. Further investigations into the biology of vascular cells in development and disease promise to have major ramifications on therapeutic strategies to combat pathologies of the vasculature.
Collapse
Affiliation(s)
- R Mazurek
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - J M Dave
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - R R Chandran
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Misra
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - A Q Sheikh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - D M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States.
| |
Collapse
|
833
|
Abstract
PURPOSE OF REVIEW Cell senescence is a major process regulating tissue mass, architecture and function, and underlies many diseases of ageing. Recent studies have elucidated some of the regulatory pathways leading to cell senescence, and senescence has also been found in the vasculature. However, assessment of cell senescence is problematic, and the effects of vascular cell senescence are in most cases unproven. The present article will review how senescence is assessed, how it is regulated, where senescence has been described, and the role of cell senescence in atherosclerosis. RECENT FINDINGS Senescence results in expression of multiple proteins, both intracellular and secreted. However, to date, none of these are specific for senescence, and multiple markers must be used together for positive identification. Despite these shortfalls, cell senescence is detectable in the vasculature in ageing and in human atherosclerosis, and recent studies in mice have indicated that cell senescence promotes both atherogenesis and multiple features of 'vulnerable' lesions in advanced atherosclerotic plaques. SUMMARY The almost ubiquitous presence of cell senescence in atherosclerosis and the fundamental role of senescence in regulating plaque development and stability suggest that prevention or amelioration of senescence in atherosclerosis is a viable therapeutic target.
Collapse
Affiliation(s)
- Abel Martin Garrido
- Division of Cardiovascular Medicine, University of Cambridge, Box 110, Addenbrooke's Centre for Clinical Investigation, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
834
|
Chappell J, Harman JL, Narasimhan VM, Yu H, Foote K, Simons BD, Bennett MR, Jørgensen HF. Extensive Proliferation of a Subset of Differentiated, yet Plastic, Medial Vascular Smooth Muscle Cells Contributes to Neointimal Formation in Mouse Injury and Atherosclerosis Models. Circ Res 2016; 119:1313-1323. [PMID: 27682618 PMCID: PMC5149073 DOI: 10.1161/circresaha.116.309799] [Citation(s) in RCA: 302] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/13/2016] [Accepted: 09/27/2016] [Indexed: 01/27/2023]
Abstract
Supplemental Digital Content is available in the text. Rationale: Vascular smooth muscle cell (VSMC) accumulation is a hallmark of atherosclerosis and vascular injury. However, fundamental aspects of proliferation and the phenotypic changes within individual VSMCs, which underlie vascular disease, remain unresolved. In particular, it is not known whether all VSMCs proliferate and display plasticity or whether individual cells can switch to multiple phenotypes. Objective: To assess whether proliferation and plasticity in disease is a general characteristic of VSMCs or a feature of a subset of cells. Methods and Results: Using multicolor lineage labeling, we demonstrate that VSMCs in injury-induced neointimal lesions and in atherosclerotic plaques are oligoclonal, derived from few expanding cells. Lineage tracing also revealed that the progeny of individual VSMCs contributes to both alpha smooth muscle actin (aSma)–positive fibrous cap and Mac3-expressing macrophage-like plaque core cells. Costaining for phenotypic markers further identified a double-positive aSma+ Mac3+ cell population, which is specific to VSMC-derived plaque cells. In contrast, VSMC-derived cells generating the neointima after vascular injury generally retained the expression of VSMC markers and the upregulation of Mac3 was less pronounced. Monochromatic regions in atherosclerotic plaques and injury-induced neointima did not contain VSMC-derived cells expressing a different fluorescent reporter protein, suggesting that proliferation-independent VSMC migration does not make a major contribution to VSMC accumulation in vascular disease. Conclusions: We demonstrate that extensive proliferation of a low proportion of highly plastic VSMCs results in the observed VSMC accumulation after injury and in atherosclerotic plaques. Therapeutic targeting of these hyperproliferating VSMCs might effectively reduce vascular disease without affecting vascular integrity.
Collapse
Affiliation(s)
- Joel Chappell
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Jennifer L Harman
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Vagheesh M Narasimhan
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Haixiang Yu
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Kirsty Foote
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Benjamin D Simons
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Martin R Bennett
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.)
| | - Helle F Jørgensen
- From the Cardiovascular Medicine Division, Department of Medicine (J.C., J.L.H., H.Y., K.F., M.R.B., H.F.J.), Cavendish Laboratory, Department of Physics (B.D.S.), The Wellcome Trust/Cancer Research UK Gurdon Institute (B.D.S.), and Wellcome Trust-Medical Research Council Stem Cell Institute (B.D.S.), University of Cambridge, United Kingdom; and The Wellcome Trust Sanger Institute, Hinxton, Cambridge, United Kingdom (V.M.N.).
| |
Collapse
|
835
|
Lin ME, Chen TM, Wallingford MC, Nguyen NB, Yamada S, Sawangmake C, Zhang J, Speer MY, Giachelli CM. Runx2 deletion in smooth muscle cells inhibits vascular osteochondrogenesis and calcification but not atherosclerotic lesion formation. Cardiovasc Res 2016; 112:606-616. [PMID: 27671804 DOI: 10.1093/cvr/cvw205] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 08/05/2016] [Indexed: 01/23/2023] Open
Abstract
Aims Vascular smooth muscle cells (SMCs) are major precursors contributing to osteochondrogenesis and calcification in atherosclerosis. Runt-related transcription factor-2 (Runx2) has been found essential for SMC differentiation to an osteochondrogenic phenotype and subsequent calcification in vitro. A recent study using a conditional targeting allele that produced a truncated Runx2 protein in SMCs of ApoE-/- mice showed reduced vascular calcification, likely occurring via reduction of receptor activator of nuclear factor-κB ligand (RANKL), macrophage infiltration, and atherosclerotic lesion formation. Using an improved conditional Runx2 knockout mouse model, we have elucidated new roles for SMC-specific Runx2 in arterial intimal calcification (AIC) without effects on atherosclerotic lesion size. Methods and results We used an improved targeting construct to generate LDLr-/- mice with floxed-Runx2 alleles ( LDLr-/- :Runx2 f/f ) such that Cre-mediated recombination ( LDLr-/- :Runx2 ΔSM ) does not produce functional truncated Runx2 protein, thereby avoiding off-target effects. We found that both LDLr-/- :Runx2 f/f and LDLr-/- :Runx2 ΔSM mice fed with a high fat diet developed atherosclerosis. SMC-specific Runx2 deletion did not significantly reduce atherosclerotic lesion size, macrophage number, or expression of RANKL, MCP-1, and CCR2. However, it significantly reduced AIC by 50%. Mechanistically, Sox9 and type II collagen were unaltered in vessels of LDLr-/- :Runx2 ΔSM mice compared to LDLr-/- :Runx2 f/f counterparts, while type X collagen, MMP13 and the osteoblastic marker osteocalcin were significantly reduced. Conclusions SMC autonomous Runx2 is required for SMC differentiation towards osteoblast-like cells, SMC-derived chondrocyte maturation and AIC in atherosclerotic mice. These effects were independent of systemic lipid metabolism, RANKL expression, macrophage infiltration, and atheromatous lesion progression.
Collapse
Affiliation(s)
- Mu-En Lin
- Present address. RevMAb Biosciences, 870 Dubuque Ave, South San Francisco, CA 94080, USA
| | | | | | - Ngoc B Nguyen
- Present address. University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Chenphop Sawangmake
- Present address. Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | | | | | | |
Collapse
|
836
|
Chen PY, Qin L, Li G, Tellides G, Simons M. Fibroblast growth factor (FGF) signaling regulates transforming growth factor beta (TGFβ)-dependent smooth muscle cell phenotype modulation. Sci Rep 2016; 6:33407. [PMID: 27634335 PMCID: PMC5025753 DOI: 10.1038/srep33407] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/24/2016] [Indexed: 12/12/2022] Open
Abstract
Smooth muscle cells (SMCs) in normal blood vessels exist in a highly differentiate state characterized by expression of SMC-specific contractile proteins ("contractile phenotype"). Following blood vessel injury in vivo or when cultured in vitro in the presence of multiple growth factors, SMC undergo a phenotype switch characterized by the loss of contractile markers and appearance of expression of non-muscle proteins ("proliferative phenotype"). While a number of factors have been reported to modulate this process, its regulation remains uncertain. Here we show that induction of SMC FGF signaling inhibits TGFβ signaling and converts contractile SMCs to the proliferative phenotype. Conversely, inhibition of SMC FGF signaling induces TGFβ signaling converting proliferating SMCs to the contractile phenotype, even in the presence of various growth factors in vitro or vascular injury in vivo. The importance of this signaling cross-talk is supported by in vivo data that show that an SMC deletion of a pan-FGF receptor adaptor Frs2α (fibroblast growth factor receptor substrate 2 alpha) in mice profoundly reduces neointima formation and vascular remodelling following carotid artery ligation. These results demonstrate that FGF-TGFβ signaling antagonism is the primary regulator of the SMC phenotype switch. Manipulation of this cross-talk may be an effective strategy for treatment of SMC-proliferation related diseases.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Guangxin Li
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Department of Vascular Surgery, The First Hospital of China Medical University, 155 Nanjing Bei Street, Shenyang, China
| | - George Tellides
- Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
837
|
Kramann R, Goettsch C, Wongboonsin J, Iwata H, Schneider RK, Kuppe C, Kaesler N, Chang-Panesso M, Machado FG, Gratwohl S, Madhurima K, Hutcheson JD, Jain S, Aikawa E, Humphreys BD. Adventitial MSC-like Cells Are Progenitors of Vascular Smooth Muscle Cells and Drive Vascular Calcification in Chronic Kidney Disease. Cell Stem Cell 2016; 19:628-642. [PMID: 27618218 DOI: 10.1016/j.stem.2016.08.001] [Citation(s) in RCA: 239] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 06/13/2016] [Accepted: 08/01/2016] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cell (MSC)-like cells reside in the vascular wall, but their role in vascular regeneration and disease is poorly understood. Here, we show that Gli1+ cells located in the arterial adventitia are progenitors of vascular smooth muscle cells and contribute to neointima formation and repair after acute injury to the femoral artery. Genetic fate tracing indicates that adventitial Gli1+ MSC-like cells migrate into the media and neointima during athero- and arteriosclerosis in ApoE-/- mice with chronic kidney disease. Our data indicate that Gli1+ cells are a major source of osteoblast-like cells during calcification in the media and intima. Genetic ablation of Gli1+ cells before induction of kidney injury dramatically reduced the severity of vascular calcification. These findings implicate Gli1+ cells as critical adventitial progenitors in vascular remodeling after acute and during chronic injury and suggest that they may be relevant therapeutic targets for mitigation of vascular calcification.
Collapse
Affiliation(s)
- Rafael Kramann
- Division of Nephrology and Clinical Immunology, Medical Faculty RWTH Aachen University, RWTH Aachen University, 52074 Aachen, Germany; Renal Division, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02138, USA.
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Janewit Wongboonsin
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rebekka K Schneider
- Division of Hematology, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA 02138, USA; Division of Hematology, RWTH Aachen University, 52074 Aachen, Germany
| | - Christoph Kuppe
- Division of Nephrology and Clinical Immunology, Medical Faculty RWTH Aachen University, RWTH Aachen University, 52074 Aachen, Germany
| | - Nadine Kaesler
- Division of Nephrology and Clinical Immunology, Medical Faculty RWTH Aachen University, RWTH Aachen University, 52074 Aachen, Germany
| | - Monica Chang-Panesso
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Flavia G Machado
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susannah Gratwohl
- Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kaushal Madhurima
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua D Hutcheson
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Center for Excellence in Vascular Biology, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02138, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
838
|
Nestin(+) cells direct inflammatory cell migration in atherosclerosis. Nat Commun 2016; 7:12706. [PMID: 27586429 PMCID: PMC5025806 DOI: 10.1038/ncomms12706] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 07/25/2016] [Indexed: 02/02/2023] Open
Abstract
Atherosclerosis is a leading death cause. Endothelial and smooth muscle cells participate in atherogenesis, but it is unclear whether other mesenchymal cells contribute to this process. Bone marrow (BM) nestin+ cells cooperate with endothelial cells in directing monocyte egress to bloodstream in response to infections. However, it remains unknown whether nestin+ cells regulate inflammatory cells in chronic inflammatory diseases, such as atherosclerosis. Here, we show that nestin+ cells direct inflammatory cell migration during chronic inflammation. In Apolipoprotein E (ApoE) knockout mice fed with high-fat diet, BM nestin+ cells regulate the egress of inflammatory monocytes and neutrophils. In the aorta, nestin+ stromal cells increase ∼30 times and contribute to the atheroma plaque. Mcp1 deletion in nestin+ cells—but not in endothelial cells only— increases circulating inflammatory cells, but decreases their aortic infiltration, delaying atheroma plaque formation and aortic valve calcification. Therefore, nestin expression marks cells that regulate inflammatory cell migration during atherosclerosis. Bone marrow cells producing the intermediate filament nestin guide monocyte egress to the bloodstream in response to infection. Here, the authors show that nestin-producing stromal cells direct inflammatory cell migration in atherosclerosis, and that stromal Mcp1 is crucial in this process.
Collapse
|
839
|
Sheikh AQ, Misra A, Rosas IO, Adams RH, Greif DM. Smooth muscle cell progenitors are primed to muscularize in pulmonary hypertension. Sci Transl Med 2016; 7:308ra159. [PMID: 26446956 DOI: 10.1126/scitranslmed.aaa9712] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Excess and ectopic smooth muscle cells (SMCs) are central to cardiovascular disease pathogenesis, but underlying mechanisms are poorly defined. For instance, pulmonary hypertension (PH) or elevated pulmonary artery blood pressure is a devastating disease with distal extension of smooth muscle to normally unmuscularized pulmonary arterioles. We identify novel SMC progenitors that are located at the pulmonary arteriole muscular-unmuscular border and express both SMC markers and the undifferentiated mesenchyme marker platelet-derived growth factor receptor-β (PDGFR-β). We term these cells "primed" because in hypoxia-induced PH, they express the pluripotency factor Kruppel-like factor 4 (KLF4), and in each arteriole, one of them migrates distally, dedifferentiates, and clonally expands, giving rise to the distal SMCs. Furthermore, hypoxia-induced expression of the ligand PDGF-B regulates primed cell KLF4 expression, and enhanced PDGF-B and KLF4 levels are required for distal arteriole muscularization and PH. Finally, in PH patients, KLF4 is markedly up-regulated in pulmonary arteriole smooth muscle, especially in proliferating SMCs. In sum, we have identified a pool of SMC progenitors that are critical for the pathogenesis of PH, and perhaps other vascular disorders, and therapeutic strategies targeting this cell type promise to have profound implications.
Collapse
Affiliation(s)
- Abdul Q Sheikh
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 773J, New Haven, CT 06511, USA
| | - Ashish Misra
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 773J, New Haven, CT 06511, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, 48149 Münster, Germany
| | - Daniel M Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, 300 George Street, Room 773J, New Haven, CT 06511, USA.
| |
Collapse
|
840
|
Sandison ME, Dempster J, McCarron JG. The transition of smooth muscle cells from a contractile to a migratory, phagocytic phenotype: direct demonstration of phenotypic modulation. J Physiol 2016; 594:6189-6209. [PMID: 27393389 PMCID: PMC5088226 DOI: 10.1113/jp272729] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/26/2016] [Indexed: 12/13/2022] Open
Abstract
Key points Smooth muscle cell (SMC) phenotypic conversion from a contractile to a migratory phenotype is proposed to underlie cardiovascular disease but its contribution to vascular remodelling and even its existence have recently been questioned. Tracking the fate of individual SMCs is difficult as no specific markers of migratory SMCs exist. This study used a novel, prolonged time‐lapse imaging approach to continuously track the behaviour of unambiguously identified, fully differentiated SMCs. In response to serum, highly‐elongated, contractile SMCs initially rounded up, before spreading and migrating and these migratory cells displayed clear phagocytic activity. This study provides a direct demonstration of the transition of fully contractile SMCs to a non‐contractile, migratory phenotype with phagocytic capacity that may act as a macrophage‐like cell.
Abstract Atherosclerotic plaques are populated with smooth muscle cells (SMCs) and macrophages. SMCs are thought to accumulate in plaques because fully differentiated, contractile SMCs reprogramme into a ‘synthetic’ migratory phenotype, so‐called phenotypic modulation, whilst plaque macrophages are thought to derive from blood‐borne myeloid cells. Recently, these views have been challenged, with reports that SMC phenotypic modulation may not occur during vascular remodelling and that plaque macrophages may not be of haematopoietic origin. Following the fate of SMCs is complicated by the lack of specific markers for the migratory phenotype and direct demonstrations of phenotypic modulation are lacking. Therefore, we employed long‐term, high‐resolution, time‐lapse microscopy to track the fate of unambiguously identified, fully‐differentiated, contractile SMCs in response to the growth factors present in serum. Phenotypic modulation was clearly observed. The highly elongated, contractile SMCs initially rounded up, for 1–3 days, before spreading outwards. Once spread, the SMCs became motile and displayed dynamic cell‐cell communication behaviours. Significantly, they also displayed clear evidence of phagocytic activity. This macrophage‐like behaviour was confirmed by their internalisation of 1 μm fluorescent latex beads. However, migratory SMCs did not uptake acetylated low‐density lipoprotein or express the classic macrophage marker CD68. These results directly demonstrate that SMCs may rapidly undergo phenotypic modulation and develop phagocytic capabilities. Resident SMCs may provide a potential source of macrophages in vascular remodelling. Smooth muscle cell (SMC) phenotypic conversion from a contractile to a migratory phenotype is proposed to underlie cardiovascular disease but its contribution to vascular remodelling and even its existence have recently been questioned. Tracking the fate of individual SMCs is difficult as no specific markers of migratory SMCs exist. This study used a novel, prolonged time‐lapse imaging approach to continuously track the behaviour of unambiguously identified, fully differentiated SMCs. In response to serum, highly‐elongated, contractile SMCs initially rounded up, before spreading and migrating and these migratory cells displayed clear phagocytic activity. This study provides a direct demonstration of the transition of fully contractile SMCs to a non‐contractile, migratory phenotype with phagocytic capacity that may act as a macrophage‐like cell.
Collapse
Affiliation(s)
- Mairi E Sandison
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - John Dempster
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK
| | - John G McCarron
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, SIPBS Building, 161 Cathedral Street, Glasgow, G4 0RE, UK.
| |
Collapse
|
841
|
Bot I, Kuiper J. The origin of atherosclerotic plaque cells: Plasticity or not? Atherosclerosis 2016; 251:536-537. [DOI: 10.1016/j.atherosclerosis.2016.06.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 01/28/2023]
|
842
|
Czepluch FS, Meier J, Binder C, Hasenfuss G, Schäfer K. CCL5 deficiency reduces neointima formation following arterial injury and thrombosis in apolipoprotein E-deficient mice. Thromb Res 2016; 144:136-43. [DOI: 10.1016/j.thromres.2016.06.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/12/2016] [Accepted: 06/14/2016] [Indexed: 01/21/2023]
|
843
|
Di Marco E, Gray SP, Kennedy K, Szyndralewiez C, Lyle AN, Lassègue B, Griendling KK, Cooper ME, Schmidt HHHW, Jandeleit-Dahm KAM. NOX4-derived reactive oxygen species limit fibrosis and inhibit proliferation of vascular smooth muscle cells in diabetic atherosclerosis. Free Radic Biol Med 2016; 97:556-567. [PMID: 27445103 PMCID: PMC5446082 DOI: 10.1016/j.freeradbiomed.2016.07.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 07/03/2016] [Accepted: 07/16/2016] [Indexed: 12/15/2022]
Abstract
Smooth muscle cell (SMC) proliferation and fibrosis contribute to the development of advanced atherosclerotic lesions. Oxidative stress caused by increased production or unphysiological location of reactive oxygen species (ROS) is a known major pathomechanism. However, in atherosclerosis, in particular under hyperglycaemic/diabetic conditions, the hydrogen peroxide-producing NADPH oxidase type 4 (NOX4) is protective. Here we aim to elucidate the mechanisms underlying this paradoxical atheroprotection of vascular smooth muscle NOX4 under conditions of normo- and hyperglycaemia both in vivo and ex vivo. Following 20-weeks of streptozotocin-induced diabetes, Apoe(-/-) mice showed a reduction in SM-alpha-actin and calponin gene expression with concomitant increases in platelet-derived growth factor (PDGF), osteopontin (OPN) and the extracellular matrix (ECM) protein fibronectin when compared to non-diabetic controls. Genetic deletion of Nox4 (Nox4(-/)(-)Apoe(-/-)) exacerbated diabetes-induced expression of PDGF, OPN, collagen I, and proliferation marker Ki67. Aortic SMCs isolated from NOX4-deficient mice exhibited a dedifferentiated phenotype including loss of contractile gene expression, increased proliferation and ECM production as well as elevated levels of NOX1-associated ROS. Mechanistic studies revealed that elevated PDGF signalling in NOX4-deficient SMCs mediated the loss of calponin and increase in fibronectin, while the upregulation of NOX1 was associated with the increased expression of OPN and markers of proliferation. These findings demonstrate that NOX4 actively regulates SMC pathophysiological responses in diabetic Apoe(-/-) mice and in primary mouse SMCs through the activities of PDGF and NOX1.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/pathology
- Atherosclerosis/enzymology
- Atherosclerosis/etiology
- Atherosclerosis/pathology
- Becaplermin
- Cell Proliferation
- Cells, Cultured
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/pathology
- Fibrosis
- Male
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/physiology
- NADPH Oxidase 1/metabolism
- NADPH Oxidase 4/genetics
- NADPH Oxidase 4/metabolism
- Osteopontin/genetics
- Osteopontin/metabolism
- Proto-Oncogene Proteins c-sis/genetics
- Proto-Oncogene Proteins c-sis/metabolism
- Reactive Oxygen Species/metabolism
- Superoxides/metabolism
Collapse
Affiliation(s)
- Elyse Di Marco
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne, Australia
| | - Stephen P Gray
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne, Australia
| | - Kit Kennedy
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | | | - Alicia N Lyle
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, USA
| | - Bernard Lassègue
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, USA
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, USA
| | - Mark E Cooper
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia
| | - Harald H H W Schmidt
- Department of Pharmacology & Cardiovascular Research Institute Maastricht (CARIM), Faculty of Medicine, Health & Life Science, Maastricht University, The Netherlands
| | - Karin A M Jandeleit-Dahm
- Diabetic Complications Division, Baker IDI Heart & Diabetes Institute, Melbourne, Australia; Department of Medicine, Monash University, Melbourne, Australia.
| |
Collapse
|
844
|
Perisic Matic L, Rykaczewska U, Razuvaev A, Sabater-Lleal M, Lengquist M, Miller CL, Ericsson I, Röhl S, Kronqvist M, Aldi S, Magné J, Paloschi V, Vesterlund M, Li Y, Jin H, Diez MG, Roy J, Baldassarre D, Veglia F, Humphries SE, de Faire U, Tremoli E, Odeberg J, Vukojević V, Lehtiö J, Maegdefessel L, Ehrenborg E, Paulsson-Berne G, Hansson GK, Lindeman JHN, Eriksson P, Quertermous T, Hamsten A, Hedin U. Phenotypic Modulation of Smooth Muscle Cells in Atherosclerosis Is Associated With Downregulation of LMOD1, SYNPO2, PDLIM7, PLN, and SYNM. Arterioscler Thromb Vasc Biol 2016; 36:1947-61. [PMID: 27470516 DOI: 10.1161/atvbaha.116.307893] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 07/12/2016] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Key augmented processes in atherosclerosis have been identified, whereas less is known about downregulated pathways. Here, we applied a systems biology approach to examine suppressed molecular signatures, with the hypothesis that they may provide insight into mechanisms contributing to plaque stability. APPROACH AND RESULTS Muscle contraction, muscle development, and actin cytoskeleton were the most downregulated pathways (false discovery rate=6.99e-21, 1.66e-6, 2.54e-10, respectively) in microarrays from human carotid plaques (n=177) versus healthy arteries (n=15). In addition to typical smooth muscle cell (SMC) markers, these pathways also encompassed cytoskeleton-related genes previously not associated with atherosclerosis. SYNPO2, SYNM, LMOD1, PDLIM7, and PLN expression positively correlated to typical SMC markers in plaques (Pearson r>0.6, P<0.0001) and in rat intimal hyperplasia (r>0.8, P<0.0001). By immunohistochemistry, the proteins were expressed in SMCs in normal vessels, but largely absent in human plaques and intimal hyperplasia. Subcellularly, most proteins localized to the cytoskeleton in cultured SMCs and were regulated by active enhancer histone modification H3K27ac by chromatin immunoprecipitation-sequencing. Functionally, the genes were downregulated by PDGFB (platelet-derived growth factor beta) and IFNg (interferron gamma), exposure to shear flow stress, and oxLDL (oxidized low-density lipoprotein) loading. Genetic variants in PDLIM7, PLN, and SYNPO2 loci associated with progression of carotid intima-media thickness in high-risk subjects without symptoms of cardiovascular disease (n=3378). By eQTL (expression quantitative trait locus), rs11746443 also associated with PDLIM7 expression in plaques. Mechanistically, silencing of PDLIM7 in vitro led to downregulation of SMC markers and disruption of the actin cytoskeleton, decreased cell spreading, and increased proliferation. CONCLUSIONS We identified a panel of genes that reflect the altered phenotype of SMCs in vascular disease and could be early sensitive markers of SMC dedifferentiation.
Collapse
Affiliation(s)
- Ljubica Perisic Matic
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.).
| | - Urszula Rykaczewska
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Anton Razuvaev
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Maria Sabater-Lleal
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Mariette Lengquist
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Clint L Miller
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Ida Ericsson
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Samuel Röhl
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Malin Kronqvist
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Silvia Aldi
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Joelle Magné
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Valentina Paloschi
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Mattias Vesterlund
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Yuhuang Li
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Hong Jin
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Maria Gonzalez Diez
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Joy Roy
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Damiano Baldassarre
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Fabrizio Veglia
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Steve E Humphries
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Ulf de Faire
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Elena Tremoli
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Jacob Odeberg
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Vladana Vukojević
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Janne Lehtiö
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Lars Maegdefessel
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Ewa Ehrenborg
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Gabrielle Paulsson-Berne
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Göran K Hansson
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Jan H N Lindeman
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Per Eriksson
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Thomas Quertermous
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Anders Hamsten
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| | - Ulf Hedin
- From the Departments of Molecular Medicine and Surgery (L.P.M., U.R., A.R., M.L., I.E., S.R., M.K., S.A., J.R., U.H.), Medicine (M.S.-L., J.M., V.P., Y.L., H.J., M.G.D., L.M., E.E., G.P.-B., G.K.H., P.E., A.H.), Division of Cardiovascular Epidemiology, Institute of Environmental Medicine (U.d.F.), and Department of Clinical Neuroscience, Center for Molecular Medicine (V.V.), Karolinska Institutet, Solna, Sweden; Division of Vascular Surgery, Stanford University, CA (C.L.M., T.Q.); Science for Life Laboratory, Solna, Sweden (M.V., J.L.); Dipartimento di Scienze Farmacologiche e Biomolecolari, Università di Milano, Italy (D.B., E.T.); Dipartimento di Scienze Cliniche e di Comunità, Centro Cardiologico Monzino, IRCCS, Milan, Italy (D.B., F.V., E.T.); British Heart Foundation Laboratories, Department of Medicine, University College of London, United Kingdom (S.E.H.); Department of Cardiology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden (U.d.F.); Science for Life Laboratory, Department of Proteomics, Stockholm, Sweden (J.O.); and Department of Vascular Surgery, Leiden University Medical Center, The Netherlands (J.H.N.L.)
| |
Collapse
|
845
|
Coll-Bonfill N, Peinado VI, Pisano MV, Párrizas M, Blanco I, Evers M, Engelmann JC, García-Lucio J, Tura-Ceide O, Meister G, Barberà JA, Musri MM. Slug Is Increased in Vascular Remodeling and Induces a Smooth Muscle Cell Proliferative Phenotype. PLoS One 2016; 11:e0159460. [PMID: 27441378 PMCID: PMC4956159 DOI: 10.1371/journal.pone.0159460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/01/2016] [Indexed: 12/04/2022] Open
Abstract
Objective Previous studies have confirmed Slug as a key player in regulating phenotypic changes in several cell models, however, its role in smooth muscle cells (SMC) has never been assessed. The purpose of this study was to evaluate the expression of Slug during the phenotypic switch of SMC in vitro and throughout the development of vascular remodeling. Methods and Results Slug expression was decreased during both cell-to-cell contact and TGFβ1 induced SMC differentiation. Tumor necrosis factor-α (TNFα), a known inductor of a proliferative/dedifferentiated SMC phenotype, induces the expression of Slug in SMC. Slug knockdown blocked TNFα-induced SMC phenotypic change and significantly reduced both SMC proliferation and migration, while its overexpression blocked the TGFβ1-induced SMC differentiation and induced proliferation and migration. Genome-wide transcriptomic analysis showed that in SMC, Slug knockdown induced changes mainly in genes related to proliferation and migration, indicating that Slug controls these processes in SMC. Notably, Slug expression was significantly up-regulated in lungs of mice using a model of pulmonary hypertension-related vascular remodeling. Highly remodeled human pulmonary arteries also showed an increase of Slug expression compared to less remodeled arteries. Conclusions Slug emerges as a key transcription factor driving SMC towards a proliferative phenotype. The increased Slug expression observed in vivo in highly remodeled arteries of mice and human suggests a role of Slug in the pathogenesis of pulmonary vascular diseases.
Collapse
Affiliation(s)
- Núria Coll-Bonfill
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Victor I. Peinado
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - María V. Pisano
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Isabel Blanco
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Maurits Evers
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Julia C. Engelmann
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Jessica García-Lucio
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Olga Tura-Ceide
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Gunter Meister
- Biochemistry Center Regensburg (BZR), Laboratory for RNA Biology, University of Regensburg, Regensburg, Germany
| | - Joan Albert Barberà
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Center on Respiratory Diseases (CIBERES), Madrid, Spain
| | - Melina M. Musri
- Department of Pulmonary Medicine, Hospital Clínic-Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
- Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
846
|
Bradfield PF, Menon A, Miljkovic-Licina M, Lee BP, Fischer N, Fish RJ, Kwak B, Fisher EA, Imhof BA. Divergent JAM-C Expression Accelerates Monocyte-Derived Cell Exit from Atherosclerotic Plaques. PLoS One 2016; 11:e0159679. [PMID: 27442505 PMCID: PMC4956249 DOI: 10.1371/journal.pone.0159679] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis, caused in part by monocytes in plaques, continues to be a disease that afflicts the modern world. Whilst significant steps have been made in treating this chronic inflammatory disease, questions remain on how to prevent monocyte and macrophage accumulation in atherosclerotic plaques. Junctional Adhesion Molecule C (JAM-C) expressed by vascular endothelium directs monocyte transendothelial migration in a unidirectional manner leading to increased inflammation. Here we show that interfering with JAM-C allows reverse-transendothelial migration of monocyte-derived cells, opening the way back out of the inflamed environment. To study the role of JAM-C in plaque regression we used a mouse model of atherosclerosis, and tested the impact of vascular JAM-C expression levels on monocyte reverse transendothelial migration using human cells. Studies in-vitro under inflammatory conditions revealed that overexpression or gene silencing of JAM-C in human endothelium exposed to flow resulted in higher rates of monocyte reverse-transendothelial migration, similar to antibody blockade. We then transplanted atherosclerotic, plaque-containing aortic arches from hyperlipidemic ApoE-/- mice into wild-type normolipidemic recipient mice. JAM-C blockade in the recipients induced greater emigration of monocyte-derived cells and further diminished the size of atherosclerotic plaques. Our findings have shown that JAM-C forms a one-way vascular barrier for leukocyte transendothelial migration only when present at homeostatic copy numbers. We have also shown that blocking JAM-C can reduce the number of atherogenic monocytes/macrophages in plaques by emigration, providing a novel therapeutic strategy for chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Paul F. Bradfield
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
- * E-mail:
| | - Arjun Menon
- Division of Cardiology, New York University Langone Medical Center, New York, New York 10016, United States of America
| | - Marijana Miljkovic-Licina
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Boris P. Lee
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Nicolas Fischer
- NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland
| | - Richard J. Fish
- Department of Genetic Medicine and Development, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva, Switzerland
| | - Brenda Kwak
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| | - Edward A. Fisher
- Division of Cardiology, New York University Langone Medical Center, New York, New York 10016, United States of America
| | - Beat A. Imhof
- Department of Pathology and Immunology, CMU, University of Geneva, 1211, rue Michel Servet 1, Geneva 4, Switzerland
| |
Collapse
|
847
|
Ding Y, Zhang M, Zhang W, Lu Q, Cai Z, Song P, Okon IS, Xiao L, Zou MH. AMP-Activated Protein Kinase Alpha 2 Deletion Induces VSMC Phenotypic Switching and Reduces Features of Atherosclerotic Plaque Stability. Circ Res 2016; 119:718-30. [PMID: 27439892 DOI: 10.1161/circresaha.116.308689] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 07/20/2016] [Indexed: 12/21/2022]
Abstract
RATIONALE AMP-activated protein kinase (AMPK) has been reported to play a protective role in atherosclerosis. However, whether AMPKα2 controls atherosclerotic plaque stability remains unknown. OBJECTIVE The aim of this study was to evaluate the impact of AMPKα2 deletion on atherosclerotic plaque stability in advanced atherosclerosis at the brachiocephalic arteries and to elucidate the underlying mechanisms. METHODS AND RESULTS Features of atherosclerotic plaque stability and the markers for contractile or synthetic vascular smooth muscle cell (VSMC) phenotypes were monitored in the brachiocephalic arteries from Apoe(-/-)AMPKα2(-/-) mice or VSMC-specific AMPKα2(-/-) mice in an Apoe(-/-) background (Apoe(-/-)AMPKα2(sm-/-)) fed Western diet for 10 weeks. We identified that Apoe(-/-)AMPKα2(-/-) mice and Apoe(-/-)AMPKα2(sm-/-) mice exhibited similar unstable plaque features, aggravated VSMC phenotypic switching, and significant upregulation of Kruppel-like factor 4 (KLF4) in the plaques located in the brachiocephalic arteries compared with those found in Apoe(-/-) and Apoe(-/-)AMPKα2(sm+/+) control mice. Pravastatin, an AMPK activator, suppressed VSMC phenotypic switching and alleviated features of atherosclerotic plaque instability in Apoe(-/-)AMPKα2(sm+/+) mice, but not in Apoe(-/-)AMPKα2(sm-/-) mice. VSMC isolated from AMPKα2(-/-) mice displayed a significant reduction of contractile proteins(smooth muscle actin-α, calponin, and SM-MHC [smooth muscle-mysion heavy chain]) in parallel with increased detection of synthetic proteins (vimentin and osteopontin) and KLF4, as observed in vivo. KLF4-specific siRNA abolished AMPKα2 deletion-induced VSMC phenotypic switching. Furthermore, pharmacological or genetic inhibition of nuclear factor-κB significantly decreased KLF4 upregulation in VSMC from AMPKα2(-/-) mice. Finally, we found that AMPKα2 deletion markedly promoted the binding of nuclear factor-κBp65 to KLF4 promoter. CONCLUSIONS This study demonstrated that AMPKα2 deletion induces VSMC phenotypic switching and promotes features of atherosclerotic plaque instability in nuclear factor-κB-KLF4-dependent manner.
Collapse
Affiliation(s)
- Ye Ding
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Miao Zhang
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Wencheng Zhang
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Qiulun Lu
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Zhejun Cai
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Ping Song
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Imoh Sunday Okon
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Lei Xiao
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.)
| | - Ming-Hui Zou
- From the Center for Molecular and Translational Medicine, Georgia State University, Atlanta (Y.D., Q.L., Z.C., P.S., I.S.O., L.X., M.-H.Z.); Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City (M.Z.); and The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, China (W.Z.).
| |
Collapse
|
848
|
CD47-blocking antibodies restore phagocytosis and prevent atherosclerosis. Nature 2016; 536:86-90. [PMID: 27437576 PMCID: PMC4980260 DOI: 10.1038/nature18935] [Citation(s) in RCA: 481] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/16/2016] [Indexed: 12/21/2022]
Abstract
Atherosclerosis is the disease process that underlies heart attack and stroke. Advanced lesions at risk of rupture are characterized by the pathological accumulation of diseased vascular cells and apoptotic cellular debris. Why these cells are not cleared remains unknown. Here we show that atherogenesis is associated with upregulation of CD47, a key anti-phagocytic molecule that is known to render malignant cells resistant to programmed cell removal, or 'efferocytosis'. We find that administration of CD47-blocking antibodies reverses this defect in efferocytosis, normalizes the clearance of diseased vascular tissue, and ameliorates atherosclerosis in multiple mouse models. Mechanistic studies implicate the pro-atherosclerotic factor TNF-α as a fundamental driver of impaired programmed cell removal, explaining why this process is compromised in vascular disease. Similar to recent observations in cancer, impaired efferocytosis appears to play a pathogenic role in cardiovascular disease, but is not a fixed defect and may represent a novel therapeutic target.
Collapse
|
849
|
Fu Y, Gao C, Liang Y, Wang M, Huang Y, Ma W, Li T, Jia Y, Yu F, Zhu W, Cui Q, Li Y, Xu Q, Wang X, Kong W. Shift of Macrophage Phenotype Due to Cartilage Oligomeric Matrix Protein Deficiency Drives Atherosclerotic Calcification. Circ Res 2016; 119:261-276. [PMID: 27151399 DOI: 10.1161/circresaha.115.308021] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 05/05/2016] [Indexed: 12/29/2022]
Abstract
RATIONALE Intimal calcification is highly correlated with atherosclerotic plaque burden, but the underlying mechanism is poorly understood. We recently reported that cartilage oligomeric matrix protein (COMP), a component of vascular extracellular matrix, is an endogenous inhibitor of vascular smooth muscle cell calcification. OBJECTIVE To investigate whether COMP affects atherosclerotic calcification. METHODS AND RESULTS ApoE(-/-)COMP(-/-) mice fed with chow diet for 12 months manifested more extensive atherosclerotic calcification in the innominate arteries than did ApoE(-/-) mice. To investigate which origins of COMP contributed to atherosclerotic calcification, bone marrow transplantation was performed between ApoE(-/-) and ApoE(-/-)COMP(-/-) mice. Enhanced calcification was observed in mice transplanted with ApoE(-/-)COMP(-/-) bone marrow compared with mice transplanted with ApoE(-/-) bone marrow, indicating that bone marrow-derived COMP may play a critical role in atherosclerotic calcification. Furthermore, microarray profiling of wild-type and COMP(-/-) macrophages revealed that COMP-deficient macrophages exerted atherogenic and osteogenic characters. Integrin β3 protein was attenuated in COMP(-/-) macrophages, and overexpression of integrin β3 inhibited the shift of macrophage phenotypes by COMP deficiency. Furthermore, adeno-associated virus 2-integrin β3 infection attenuated atherosclerotic calcification in ApoE(-/-)COMP(-/-) mice. Mechanistically, COMP bound directly to β-tail domain of integrin β3 via its C-terminus, and blocking of the COMP-integrin β3 association by β-tail domain mimicked the COMP deficiency-induced shift in macrophage phenotypes. Similar to COMP deficiency in mice, transduction of adeno-associated virus 2-β-tail domain enhanced atherosclerotic calcification in ApoE(-/-) mice. CONCLUSIONS These results reveal that COMP deficiency acted via integrin β3 to drive macrophages toward the atherogenic and osteogenic phenotype and thereby aggravate atherosclerotic calcification.
Collapse
Affiliation(s)
- Yi Fu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Cheng Gao
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Ying Liang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Meili Wang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yaqian Huang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Wei Ma
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Tuoyi Li
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yiting Jia
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Fang Yu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Wanlin Zhu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Qinghua Cui
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Yanhui Li
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Qingbo Xu
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Xian Wang
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.)
| | - Wei Kong
- From the Department of Physiology and Pathophysiology (Y.F., C.G., Y.L., M.W., Y.H., T.L., Y.J., F.Y., X.W., W.K.), Department of Biomedical Informatics (W.M., Q.C.), Institute of Cardiovascular Sciences, School of Basic Medical Sciences (Y.L.), Peking University, Beijing, P. R. China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P. R. China (Y.F., C.G., Y.L., M.W., Y.H., W.M., T.L., Y.J., F.Y., Q.C., Y.L., X.W., W.K.); School of Biological Science and Medical Engineering, International Research Institute for Multidisciplinary Science, Beihang University, Beijing, P. R. China (W.Z.); and Cardiovascular Division, King's College London BHF Centre, London, United Kingdom (Q.X.).
| |
Collapse
|
850
|
Integrative functional genomics identifies regulatory mechanisms at coronary artery disease loci. Nat Commun 2016; 7:12092. [PMID: 27386823 PMCID: PMC4941121 DOI: 10.1038/ncomms12092] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/28/2016] [Indexed: 12/22/2022] Open
Abstract
Coronary artery disease (CAD) is the leading cause of mortality and morbidity, driven by both genetic and environmental risk factors. Meta-analyses of genome-wide association studies have identified >150 loci associated with CAD and myocardial infarction susceptibility in humans. A majority of these variants reside in non-coding regions and are co-inherited with hundreds of candidate regulatory variants, presenting a challenge to elucidate their functions. Herein, we use integrative genomic, epigenomic and transcriptomic profiling of perturbed human coronary artery smooth muscle cells and tissues to begin to identify causal regulatory variation and mechanisms responsible for CAD associations. Using these genome-wide maps, we prioritize 64 candidate variants and perform allele-specific binding and expression analyses at seven top candidate loci: 9p21.3, SMAD3, PDGFD, IL6R, BMP1, CCDC97/TGFB1 and LMOD1. We validate our findings in expression quantitative trait loci cohorts, which together reveal new links between CAD associations and regulatory function in the appropriate disease context. Coronary heart disease is the leading cause of death worldwide with multiple environmental and genetic risk factors. Here the authors integrate genomic, epigenomic and transcriptomic mapping to elucidate causal variation and mechanisms of known genetic associations.
Collapse
|