51
|
Gupta RM, Schnitzler GR, Fang S, Lee-Kim VS, Barry A. Multiomic Analysis and CRISPR Perturbation Screens Identify Endothelial Cell Programs and Novel Therapeutic Targets for Coronary Artery Disease. Arterioscler Thromb Vasc Biol 2023; 43:600-608. [PMID: 36994731 PMCID: PMC10170398 DOI: 10.1161/atvbaha.123.318328] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/01/2023] [Indexed: 03/31/2023]
Abstract
Endothelial cells (EC) are an important mediator of atherosclerosis and vascular disease. Their exposure to atherogenic risk factors such as hypertension and serum cholesterol leads to endothelial dysfunction and many disease-associated processes. Identifying which of these multiple EC functions is causally related to disease risk has been challenging. There is evidence from in vivo models and human sequencing studies that dysregulation of nitric oxide production directly affects risk of coronary artery disease. Human genetics can help prioritize the other EC functions with causal relationships because germline mutations are acquired at birth and serve as a randomized test of which pathways affect disease risk. Though several coronary artery disease risk variants have been linked to EC function, this process has been slow and laborious. Unbiased analyses of EC dysfunction using multiomic approaches promise to identify the causal genetic mechanisms responsible for vascular disease. Here, we review the data from genomic, epigenomic, and transcriptomic studies that prioritize EC-specific causal pathways. New methods that CRISPR (clustered regularly interspaced short palindromic repeats) perturbation technology with genomic, epigenomic, and transcriptomic analysis promise to speed up the characterization of disease-associated genetic variation. We summarize several recent studies in ECs which use high-throughput genetic perturbation to identify disease-relevant pathways and novel mechanisms of disease. These genetically validated pathways can accelerate the identification of drug targets for the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Rajat M Gupta
- Divisions of Genetics and Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
- Broad Institute of MIT and Harvard, Cambridge, MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
| | - Gavin R Schnitzler
- Divisions of Genetics and Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
- Broad Institute of MIT and Harvard, Cambridge, MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
| | - Shi Fang
- Divisions of Genetics and Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
- Broad Institute of MIT and Harvard, Cambridge, MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
| | - Vivian S Lee-Kim
- Divisions of Genetics and Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
- Broad Institute of MIT and Harvard, Cambridge, MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
| | - Aurelie Barry
- Divisions of Genetics and Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
- Broad Institute of MIT and Harvard, Cambridge, MA (R.M.G., G.R.S., S.F., V.S.L.-K., A.B.)
| |
Collapse
|
52
|
Yang X, Yang C, Friesel RE, Liaw L. Sprouty1 has a protective role in atherogenesis and modifies the migratory and inflammatory phenotype of vascular smooth muscle cells. Atherosclerosis 2023; 373:17-28. [PMID: 37121163 PMCID: PMC10225353 DOI: 10.1016/j.atherosclerosis.2023.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/17/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND AND AIMS Sprouty1 (Spry1) regulates the differentiation of vascular smooth muscle cells (VSMC), and our aim was to determine its role in atherogenesis. A significant proportion of cells within atherosclerotic lesions are derived from migration and pathological adaptation of medial VSMC. METHODS We used global Spry1 null mouse, and Myh11-CreERT2, ROSA26-STOPfl/fl-tdTomato-Spry1fl/fl mice to allow for lineage tracing and conditional Spry1 deletion in VSMC. Atherosclerosis was induced by injection of a mutant form of mPCSK9D377Y-AAV followed by Western diet. Human aortic VSMC (hVSMC) with shRNA targeting of Spry1 were also analyzed. RESULTS Global loss of Spry1 increased inflammatory markers ICAM1 and Cox2 in VSMC. Conditional deletion of Spry1 in VSMC had no effect on early lesion development, despite increased Sca1high cells. After 26 weeks of Western diet, mice with VSMC deletion of Spry1 had increased plaque burden, with reduced collagen content and smooth muscle alpha actin (SMA) in the fibrous cap. Lineage tracing via tdTomato marking Cre-recombined cells indicated that VSMC with loss of Spry1 had decreased migration into the lesion, noted by decreased proportions of tdTomato+ and tdTomato+/SMA + cells. Loss-of-function of Spry1 in hVSMC increased mesenchymal and activation markers, including KLF4, PDGFRb, ICAM1, and Cox2. Loss of Spry1 enhanced the effects of PDGFBB and TNFa on hVSMC. CONCLUSIONS Loss of Spry1 in VSMC aggravated plaque formation at later stages, and increased markers of instability. Our results indicate that Spry1 suppresses the mesenchymal and inflammatory phenotype of VSMC, and its expression in VSMC is protective against chronic atherosclerotic disease.
Collapse
Affiliation(s)
- Xuehui Yang
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, 04074, USA
| | - Chenhao Yang
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, USA
| | - Robert E Friesel
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, 04074, USA
| | - Lucy Liaw
- Center for Molecular Medicine, MaineHealth Institute for Research, MaineHealth, Scarborough, ME, 04074, USA; Graduate School of Biomedical Science and Engineering, University of Maine, USA.
| |
Collapse
|
53
|
Raju S, Botts SR, Blaser M, Prajapati K, Ho TWW, Ching C, Galant NJ, Fiddes L, Wu R, Clift CL, Pham T, Lee WL, Singh SA, Aikawa E, Fish JE, Howe KL. Endothelial cells secrete small extracellular vesicles bidirectionally containing distinct cargo to uniquely reprogram vascular cells in the circulation and vessel wall. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538787. [PMID: 37162986 PMCID: PMC10168399 DOI: 10.1101/2023.04.28.538787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Rationale Extracellular vesicles (EVs) contain bioactive cargo including microRNAs (miRNAs) and proteins that are released by cells as a form of cell-cell communication. Endothelial cells (ECs) form the innermost lining of all blood vessels and thereby interface with cells in the circulation as well as cells residing in the vascular wall. It is unknown whether ECs have the capacity to release EVs capable of governing recipient cells within two separate compartments, and how this is affected by endothelial activation commonly seen in atheroprone regions. Objective Given their boundary location, we propose that ECs utilize bidirectional release of distinct EV cargo in quiescent and activated states to communicate with cells within the circulation and blood vessel wall. Methods and Results EVs were isolated from primary human aortic endothelial cells (ECs) (+/-IL-1β activation), quantified, and analysed by miRNA transcriptomics and proteomics. Compared to quiescent ECs, activated ECs increased EV release, with miRNA and protein cargo that were related to atherosclerosis. RNA sequencing of EV-treated monocytes and smooth muscle cells (SMCs) revealed that EVs from activated ECs altered pathways that were pro-inflammatory and atherogenic. Apical and basolateral EV release was assessed using ECs on transwells. ECs released more EVs apically, which increased with activation. Apical and basolateral EV cargo contained distinct transcriptomes and proteomes that were altered by EC activation. Notably, basolateral EC-EVs displayed greater changes in the EV secretome, with pathways specific to atherosclerosis. In silico analysis determined that compartment-specific cargo released by the apical and basolateral surfaces of ECs can reprogram monocytes and SMCs, respectively. Conclusions The demonstration that ECs are capable of polarized EV cargo loading and directional EV secretion reveals a novel paradigm for endothelial communication, which may ultimately enhance our ability to design endothelial-based therapeutics for cardiovascular diseases such as atherosclerosis where ECs are persistently activated.
Collapse
Affiliation(s)
- Sneha Raju
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Vascular Surgery, Toronto General Hospital, Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - Mark Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kamalben Prajapati
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Tse Wing Winnie Ho
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Crizza Ching
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
| | | | - Lindsey Fiddes
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
| | - Ruilin Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Cassandra L. Clift
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tan Pham
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON, Canada
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason E Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, Canada
| | - Kathryn L Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
- Institute of Medical Science, University of Toronto, Toronto, Canada
- Division of Vascular Surgery, Toronto General Hospital, Toronto, Canada
- Faculty of Medicine, University of Toronto, Toronto ON, Canada
- Peter Munk Cardiac Centre, Toronto General Hospital, Toronto, Canada
| |
Collapse
|
54
|
de Winther MPJ, Bäck M, Evans P, Gomez D, Goncalves I, Jørgensen HF, Koenen RR, Lutgens E, Norata GD, Osto E, Dib L, Simons M, Stellos K, Ylä-Herttuala S, Winkels H, Bochaton-Piallat ML, Monaco C. Translational opportunities of single-cell biology in atherosclerosis. Eur Heart J 2023; 44:1216-1230. [PMID: 36478058 PMCID: PMC10120164 DOI: 10.1093/eurheartj/ehac686] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 12/12/2022] Open
Abstract
The advent of single-cell biology opens a new chapter for understanding human biological processes and for diagnosing, monitoring, and treating disease. This revolution now reaches the field of cardiovascular disease (CVD). New technologies to interrogate CVD samples at single-cell resolution are allowing the identification of novel cell communities that are important in shaping disease development and direct towards new therapeutic strategies. These approaches have begun to revolutionize atherosclerosis pathology and redraw our understanding of disease development. This review discusses the state-of-the-art of single-cell analysis of atherosclerotic plaques, with a particular focus on human lesions, and presents the current resolution of cellular subpopulations and their heterogeneity and plasticity in relation to clinically relevant features. Opportunities and pitfalls of current technologies as well as the clinical impact of single-cell technologies in CVD patient care are highlighted, advocating for multidisciplinary and international collaborative efforts to join the cellular dots of CVD.
Collapse
Affiliation(s)
- Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC location University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Magnus Bäck
- Translational Cardiology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- University of Lorraine, INSERM U1116, Nancy University Hospital, Nancy, France
| | - Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, INSIGNEO Institute, and the Bateson Centre, University of Sheffield, Sheffield, UK
| | - Delphine Gomez
- Department of Medicine, Division of Cardiology, Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Isabel Goncalves
- Cardiovascular Research Translational Studies, Clinical Sciences, Lund University, Malmö, Sweden
- Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - Helle F Jørgensen
- Cardiorespiratory Medicine Section, Department of Medicine, University of Cambridge, Hills Road, Cambridge CB2 0QQ, UK
| | - Rory R Koenen
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Esther Lutgens
- Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilian’s Universität, Munich, Germany
- German Centre of Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Cardiovascular Medicine, Experimental CardioVascular Immunology Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Giuseppe Danilo Norata
- Department of Excellence in Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
- Center for the Study of Atherosclerosis, SISA, Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Elena Osto
- Institute of Clinical Chemistry and Department of Cardiology, Heart Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Lea Dib
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, OX37FY Oxford, UK
| | - Michael Simons
- Departments of Internal Medicine and Cell Biology, Yale University and Yale Cardiovascular Research Center, 300 George St, New Haven, CT 06511, USA
| | - Konstantinos Stellos
- European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute, University of Eastern Finland and Heart Center, Kuopio University Hospital, Kuopio, Finland
| | - Holger Winkels
- Department of Internal Medicine III, Division of Cardiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | | | - Claudia Monaco
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, OX37FY Oxford, UK
| |
Collapse
|
55
|
Bonacina F, Di Costanzo A, Genkel V, Kong XY, Kroon J, Stimjanin E, Tsiantoulas D, Grootaert MO. The heterogeneous cellular landscape of atherosclerosis: Implications for future research and therapies. A collaborative review from the EAS young fellows. Atherosclerosis 2023; 372:48-56. [PMID: 37030081 DOI: 10.1016/j.atherosclerosis.2023.03.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/10/2023]
Abstract
Single cell technologies, lineage tracing mouse models and advanced imaging techniques unequivocally improved the resolution of the cellular landscape of atherosclerosis. Although the discovery of the heterogeneous nature of the cellular plaque architecture has undoubtedly improved our understanding of the specific cellular states in atherosclerosis progression, it also adds more complexity to current and future research and will change how we approach future drug development. In this review, we will discuss how the revolution of new single cell technologies allowed us to map the cellular networks in the plaque, but we will also address current (technological) limitations that confine us to identify the cellular drivers of the disease and to pinpoint a specific cell state, cell subset or cell surface antigen as new candidate drug target for atherosclerosis.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Alessia Di Costanzo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Vadim Genkel
- Department of Internal Medicine, South-Ural State Medical University, Chelyabinsk, Russia
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jeffrey Kroon
- Amsterdam UMC Location University of Amsterdam, Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands; Amsterdam Cardiovascular Sciences, Atherosclerosis & Ischemic Syndromes, Amsterdam, Netherlands; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, VIB, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Belgium
| | - Ena Stimjanin
- Department of Internal Medicine, Cantonal Hospital Zenical, Zenica, Bosnia and Herzegovina
| | | | - Mandy Oj Grootaert
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
56
|
Dong Y, Wang B, Du M, Zhu B, Cui K, Li K, Yuan K, Cowan DB, Bhattacharjee S, Wong S, Shi J, Wang DZ, Chen K, Bischoff J, Linton MF, Chen H. Targeting Epsins to Inhibit Fibroblast Growth Factor Signaling While Potentiating Transforming Growth Factor-β Signaling Constrains Endothelial-to-Mesenchymal Transition in Atherosclerosis. Circulation 2023; 147:669-685. [PMID: 36591786 PMCID: PMC10136057 DOI: 10.1161/circulationaha.122.063075] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/29/2022] [Indexed: 01/03/2023]
Abstract
BACKGROUND Epsin endocytic adaptor proteins are implicated in the progression of atherosclerosis; however, the underlying molecular mechanisms have not yet been fully defined. In this study, we determined how epsins enhance endothelial-to-mesenchymal transition (EndoMT) in atherosclerosis and assessed the efficacy of a therapeutic peptide in a preclinical model of this disease. METHODS Using single-cell RNA sequencing combined with molecular, cellular, and biochemical analyses, we investigated the role of epsins in stimulating EndoMT using knockout in Apoe-/- and lineage tracing/proprotein convertase subtilisin/kexin type 9 serine protease mutant viral-induced atherosclerotic mouse models. The therapeutic efficacy of a synthetic peptide targeting atherosclerotic plaques was then assessed in Apoe-/- mice. RESULTS Single-cell RNA sequencing and lineage tracing revealed that epsins 1 and 2 promote EndoMT and that the loss of endothelial epsins inhibits EndoMT marker expression and transforming growth factor-β signaling in vitro and in atherosclerotic mice, which is associated with smaller lesions in the Apoe-/- mouse model. Mechanistically, the loss of endothelial cell epsins results in increased fibroblast growth factor receptor-1 expression, which inhibits transforming growth factor-β signaling and EndoMT. Epsins directly bind ubiquitinated fibroblast growth factor receptor-1 through their ubiquitin-interacting motif, which results in endocytosis and degradation of this receptor complex. Consequently, administration of a synthetic ubiquitin-interacting motif-containing peptide atheroma ubiquitin-interacting motif peptide inhibitor significantly attenuates EndoMT and progression of atherosclerosis. CONCLUSIONS We conclude that epsins potentiate EndoMT during atherogenesis by increasing transforming growth factor-β signaling through fibroblast growth factor receptor-1 internalization and degradation. Inhibition of EndoMT by reducing epsin-fibroblast growth factor receptor-1 interaction with a therapeutic peptide may represent a novel treatment strategy for atherosclerosis.
Collapse
Affiliation(s)
- Yunzhou Dong
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Beibei Wang
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Mulong Du
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02115
| | - Bo Zhu
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Kui Cui
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Kathryn Li
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
| | - Ke Yuan
- Department of Medicine, Boston Children’s Hospital, Boston, MA 02115
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
| | - Douglas B. Cowan
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Sudarshan Bhattacharjee
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - Scott Wong
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA, 02115
- Department of Anæsthesia, Harvard Medical School, Boston, MA 02115
| | - Da-Zhi Wang
- USF Heart Institute, Center for Regenerative Medicine, University of South Florida, Tampa, FL 33612
| | - Kaifu Chen
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115
- Department of Cardiology, Boston Children’s Hospital, Boston, MA 02115
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| | - MacRae F. Linton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Hong Chen
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115
- Department of Surgery, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
57
|
Kim HJ, Cheng P, Travisano S, Weldy C, Monteiro JP, Kundu R, Nguyen T, Sharma D, Shi H, Lin Y, Liu B, Haldar S, Jackson S, Quertermous T. Molecular mechanisms of coronary artery disease risk at the PDGFD locus. Nat Commun 2023; 14:847. [PMID: 36792607 PMCID: PMC9932166 DOI: 10.1038/s41467-023-36518-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Genome wide association studies for coronary artery disease (CAD) have identified a risk locus at 11q22.3. Here, we verify with mechanistic studies that rs2019090 and PDGFD represent the functional variant and gene at this locus. Further, FOXC1/C2 transcription factor binding at rs2019090 is shown to promote PDGFD transcription through the CAD promoting allele. With single cell transcriptomic and histology studies with Pdgfd knockdown in an SMC lineage tracing male atherosclerosis mouse model we find that Pdgfd promotes expansion, migration, and transition of SMC lineage cells to the chondromyocyte phenotype. Pdgfd also increases adventitial fibroblast and pericyte expression of chemokines and leukocyte adhesion molecules, which is linked to plaque macrophage recruitment. Despite these changes there is no effect of Pdgfd deletion on overall plaque burden. These findings suggest that PDGFD mediates CAD risk by promoting deleterious phenotypic changes in SMC, along with an inflammatory response that is primarily focused in the adventitia.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Paul Cheng
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Stanislao Travisano
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Chad Weldy
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - João P Monteiro
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Ramendra Kundu
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Trieu Nguyen
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Disha Sharma
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Huitong Shi
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA
| | - Yi Lin
- Research Center for Intelligent Computing Platforms, Zhejiang Laboratory, Hangzhou, 311121, China
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Boxiang Liu
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Saptarsi Haldar
- Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Simon Jackson
- Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA, 94080, USA
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA, 94305, USA.
| |
Collapse
|
58
|
Benavente ED, Karnewar S, Buono M, Mili E, Hartman RJG, Kapteijn D, Slenders L, Daniels M, Aherrahrou R, Reinberger T, Mol BM, de Borst GJ, de Kleijn DPV, Prange KHM, Depuydt MAC, de Winther MPJ, Kuiper J, Björkegren JLM, Erdmann J, Civelek M, Mokry M, Owens GK, Pasterkamp G, den Ruijter HM. Female gene networks are expressed in myofibroblast-like smooth muscle cells in vulnerable atherosclerotic plaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.08.527690. [PMID: 36798294 PMCID: PMC9934638 DOI: 10.1101/2023.02.08.527690] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Women presenting with coronary artery disease (CAD) more often present with fibrous atherosclerotic plaques, which are currently understudied. Phenotypically modulated smooth muscle cells (SMCs) contribute to atherosclerosis in women. How these phenotypically modulated SMCs shape female versus male plaques is unknown. Here, we show sex-stratified gene regulatory networks (GRNs) from human carotid atherosclerotic tissue. Prioritization of these networks identified two main SMC GRNs in late-stage atherosclerosis. Single-cell RNA-sequencing mapped these GRNs to two SMC phenotypes: a phenotypically modulated myofibroblast-like SMC network and a contractile SMC network. The myofibroblast-like GRN was mostly expressed in plaques that were vulnerable in females. Finally, mice orthologs of the female myofibroblast-like genes showed retained expression in advanced plaques from female mice but were downregulated in male mice during atherosclerosis progression. Female atherosclerosis is driven by GRNs that promote a fibrous vulnerable plaque rich in myofibroblast-like SMCs.
Collapse
Affiliation(s)
- Ernest Diez Benavente
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Santosh Karnewar
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Michele Buono
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Eloi Mili
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Robin J. G. Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Daniek Kapteijn
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Lotte Slenders
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mark Daniels
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Redouane Aherrahrou
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Finland
| | | | - Barend M. Mol
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, Utrecht University, The Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, Utrecht University, The Netherlands
| | - Dominique P. V. de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, Utrecht University, The Netherlands
| | - Koen H. M. Prange
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Marie A. C. Depuydt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Menno P. J. de Winther
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University’ Leiden, The Netherlands
| | - Johan L. M. Björkegren
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Michal Mokry
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht University, the Netherlands
| |
Collapse
|
59
|
Deaton RA, Bulut G, Serbulea V, Salamon A, Shankman LS, Nguyen AT, Owens GK. A New Autosomal Myh11-CreERT2 Smooth Muscle Cell Lineage Tracing and Gene Knockout Mouse Model-Brief Report. Arterioscler Thromb Vasc Biol 2023; 43:203-211. [PMID: 36519470 PMCID: PMC9877184 DOI: 10.1161/atvbaha.122.318160] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The Myh11 promoter is extensively used as a smooth muscle cell (SMC) Cre-driver and is regarded as the most restrictive and specific promoter available to study SMCs. Unfortunately, in the existing Myh11-CreERT2 mouse, the transgene was inserted on the Y chromosome precluding the study of female mice. Given the importance of including sex as a biological variable and that numerous SMC-based diseases have a sex-dependent bias, the field has been tremendously limited by the lack of a model to study both sexes. Here, we describe a new autosomal Myh11-CreERT2 mouse (referred to as Myh11-CreERT2-RAD), which allows for SMC-specific lineage tracing and gene knockout studies in vivo using both male and female mice. METHODS A Myh11-CreERT2-RAD transgenic C57BL/6 mouse line was generated using bacterial artificial chromosome clone RP23-151J22 modified to contain a Cre-ERT2 after the Myh11 start codon. Myh11-CreERT2-RAD mice were crossed with 2 different fluorescent reporter mice and tested for SMC-specific labeling by flow cytometric and immunofluorescence analyses. RESULTS Myh11-CreERT2-RAD transgene insertion was determined to be on mouse chromosome 2. Myh11-CreERT2-RAD fluorescent reporter mice showed Cre-dependent, tamoxifen-inducible labeling of SMCs equivalent to the widely used Myh11-CreERT2 mice. Labeling was equivalent in both male and female Cre+ mice and was limited to vascular and visceral SMCs and pericytes in various tissues as assessed by immunofluorescence. CONCLUSIONS We generated and validated the function of an autosomal Myh11-CreERT2-RAD mouse that can be used to assess sex as a biological variable with respect to the normal and pathophysiological functions of SMCs.
Collapse
Affiliation(s)
- Rebecca A. Deaton
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Gamze Bulut
- College of William and Mary, Williamsburg, Virginia 23185
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Anita Salamon
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | - Laura S. Shankman
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| | | | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia-School of Medicine, 415 Lane Road, Suite 1010, Charlottesville, VA, 22908
| |
Collapse
|
60
|
Canfrán-Duque A, Rotllan N, Zhang X, Andrés-Blasco I, Thompson BM, Sun J, Price NL, Fernández-Fuertes M, Fowler JW, Gómez-Coronado D, Sessa WC, Giannarelli C, Schneider RJ, Tellides G, McDonald JG, Fernández-Hernando C, Suárez Y. Macrophage-Derived 25-Hydroxycholesterol Promotes Vascular Inflammation, Atherogenesis, and Lesion Remodeling. Circulation 2023; 147:388-408. [PMID: 36416142 PMCID: PMC9892282 DOI: 10.1161/circulationaha.122.059062] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 10/20/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cross-talk between sterol metabolism and inflammatory pathways has been demonstrated to significantly affect the development of atherosclerosis. Cholesterol biosynthetic intermediates and derivatives are increasingly recognized as key immune regulators of macrophages in response to innate immune activation and lipid overloading. 25-Hydroxycholesterol (25-HC) is produced as an oxidation product of cholesterol by the enzyme cholesterol 25-hydroxylase (CH25H) and belongs to a family of bioactive cholesterol derivatives produced by cells in response to fluctuating cholesterol levels and immune activation. Despite the major role of 25-HC as a mediator of innate and adaptive immune responses, its contribution during the progression of atherosclerosis remains unclear. METHODS The levels of 25-HC were analyzed by liquid chromatography-mass spectrometry, and the expression of CH25H in different macrophage populations of human or mouse atherosclerotic plaques, respectively. The effect of CH25H on atherosclerosis progression was analyzed by bone marrow adoptive transfer of cells from wild-type or Ch25h-/- mice to lethally irradiated Ldlr-/- mice, followed by a Western diet feeding for 12 weeks. Lipidomic, transcriptomic analysis and effects on macrophage function and signaling were analyzed in vitro from lipid-loaded macrophage isolated from Ldlr-/- or Ch25h-/-;Ldlr-/- mice. The contribution of secreted 25-HC to fibrous cap formation was analyzed using a smooth muscle cell lineage-tracing mouse model, Myh11ERT2CREmT/mG;Ldlr-/-, adoptively transferred with wild-type or Ch25h-/- mice bone marrow followed by 12 weeks of Western diet feeding. RESULTS We found that 25-HC accumulated in human coronary atherosclerotic lesions and that macrophage-derived 25-HC accelerated atherosclerosis progression, promoting plaque instability through autocrine and paracrine actions. 25-HC amplified the inflammatory response of lipid-loaded macrophages and inhibited the migration of smooth muscle cells within the plaque. 25-HC intensified inflammatory responses of lipid-laden macrophages by modifying the pool of accessible cholesterol in the plasma membrane, which altered Toll-like receptor 4 signaling, promoted nuclear factor-κB-mediated proinflammatory gene expression, and increased apoptosis susceptibility. These effects were independent of 25-HC-mediated modulation of liver X receptor or SREBP (sterol regulatory element-binding protein) transcriptional activity. CONCLUSIONS Production of 25-HC by activated macrophages amplifies their inflammatory phenotype, thus promoting atherogenesis.
Collapse
Affiliation(s)
- Alberto Canfrán-Duque
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Noemi Rotllan
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xinbo Zhang
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Irene Andrés-Blasco
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
- Genomics and Diabetes Unit, Health Research Institute Clinic Hospital of Valencia (INCLIVA), Valencia, Spain
| | - Bonne M Thompson
- Center for Human Nutrition. University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Sun
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nathan L Price
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Marta Fernández-Fuertes
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joseph W. Fowler
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology Yale University School of Medicine, New Haven, Connecticut, USA
| | - Diego Gómez-Coronado
- Servicio Bioquímica-Investigación, Hospital Universitario Ramón y Cajal, IRyCIS, Madrid, and CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Spain
| | - William C. Sessa
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chiara Giannarelli
- Department of Medicine, Cardiology, NYU Grossman School of Medicine, New York, New York, USA
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - Robert J Schneider
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - George Tellides
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Surgery, Yale University School of Medicine, New Haven, Connecticut, 06520 USA
| | - Jeffrey G McDonald
- Center for Human Nutrition. University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology. Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Comparative Medicine. Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology. Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
61
|
Kim HJ, Cheng P, Travisano S, Weldy C, Monteiro JP, Kundu R, Nguyen T, Sharma D, Shi H, Lin Y, Liu B, Haldar S, Jackson S, Quertermous T. Molecular mechanisms of coronary artery disease risk at the PDGFD locus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525789. [PMID: 36747745 PMCID: PMC9900883 DOI: 10.1101/2023.01.26.525789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Platelet derived growth factor (PDGF) signaling has been extensively studied in the context of vascular disease, but the genetics of this pathway remain to be established. Genome wide association studies (GWAS) for coronary artery disease (CAD) have identified a risk locus at 11q22.3, and we have verified with fine mapping approaches that the regulatory variant rs2019090 and PDGFD represent the functional variant and putative functional gene. Further, FOXC1/C2 transcription factor (TF) binding at rs2019090 was found to promote PDGFD transcription through the CAD promoting allele. Employing a constitutive Pdgfd knockout allele along with SMC lineage tracing in a male atherosclerosis mouse model we mapped single cell transcriptomic, cell state, and lesion anatomical changes associated with gene loss. These studies revealed that Pdgfd promotes expansion, migration, and transition of SMC lineage cells to the chondromyocyte phenotype and vascular calcification. This is in contrast to protective CAD genes TCF21, ZEB2, and SMAD3 which we have shown to promote the fibroblast-like cell transition or perturb the pattern or extent of transition to the chondromyocyte phenotype. Further, Pdgfd expressing fibroblasts and pericytes exhibited greater expression of chemokines and leukocyte adhesion molecules, consistent with observed increased macrophage recruitment to the plaque. Despite these changes there was no effect of Pdgfd deletion on SMC contribution to the fibrous cap or overall lesion burden. These findings suggest that PDGFD mediates CAD risk through promoting SMC expansion and migration, in conjunction with deleterious phenotypic changes, and through promoting an inflammatory response that is primarily focused in the adventitia where it contributes to leukocyte trafficking to the diseased vessel wall.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Paul Cheng
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Stanislao Travisano
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Chad Weldy
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - João P. Monteiro
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Ramendra Kundu
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Trieu Nguyen
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Disha Sharma
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Huitong Shi
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| | - Yi Lin
- Research Center for Intelligent Computing Platforms, Zhejiang Laboratory, China 311121
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 117543
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Boxiang Liu
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore 117543
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228
| | - Saptarsi Haldar
- Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA 94080
| | - Simon Jackson
- Amgen Inc., 1120 Veterans Blvd, South San Francisco, CA 94080
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, 300 Pasteur Drive, Falk CVRC, Stanford, CA; 94305
| |
Collapse
|
62
|
Bian S, Jiang Y, Dai Z, Wu X, Li B, Wang N, Bian W, Zhong W. Lin28b delays vasculature aging by reducing platelet-derived growth factor-beta resistance in senescent vascular smooth muscle cells. Atherosclerosis 2023; 364:29-38. [PMID: 36529087 DOI: 10.1016/j.atherosclerosis.2022.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/12/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Platelet-derived growth factor-β (PDGFB) is an important mediator of vascular smooth muscle cell (VSMC) proliferation, and PDGFB resistance is observed in senescent VSMCs. Lin28b is a stemness regulator in the embryo; however, its role in vasculature aging and VSMC senescence is unknown. We aimed to investigate whether Lin28b could restore the VSMC response to PDGFB and delay vasculature aging. METHODS ApoE-/- mice were fed a high-fat diet for different weeks to establish an aging model. PDGFB resistance was observed using EdU staining in vessel culture in vitro. Quantitative polymerase chain reaction and in situ hybridization were used to detect let-7 expression. Senescence was identified by Western blotting, senescence-associated beta-galactosidase activity or Sudan Black B staining, and VSMC function was determined using CCK-8, migration, and enzyme-linked immunosorbent assays. RESULTS Vessels from aged mice showed poor responses to PDGFB stimulation compared with those from young mice; similar results were found in senescent VSMCs. The expression levels of Lin28b and PDGF receptor-β were downregulated in aging vasculature and senescent VSMCs, whereas let-7 family levels increased with aging and VSMC passage growth. Transfection of VSMCs with let-7c induced PDGFB resistance and accelerated VSMC senescence, whereas blocking let-7c restored PDGFB reactions in VSMCs. Overexpression of Lin28b protein by lentivirus resulted in the restoration of PDGFB reactions and delayed VSMC senescence, which was blocked by a let-7c mimic. CONCLUSIONS This study reveals the role of Lin28b in delaying vasculature aging by decreasing senescent VSMC PDGFB resistance mediated by let-7.
Collapse
Affiliation(s)
- Shihui Bian
- Department of Geriatrics, Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Yu Jiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Zhiyin Dai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Xi Wu
- Department of Geriatrics, Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Bo Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Nan Wang
- Department of Geriatrics, Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Wenyan Bian
- Department of Geriatrics, Affiliated Renmin Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China
| | - Wei Zhong
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, PR China.
| |
Collapse
|
63
|
Martín-Vañó S, Miralles-Abella A, Castaño P, Hurtado-Genovés G, Aguilar-Ballester M, Herrero-Cervera A, Vinué A, Martínez-Hervás S, González-Navarro H. Vascular smooth muscle cell phenotype is modulated by ligands of the lymphotoxin β receptor and the tumor necrosis factor receptor. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE ARTERIOSCLEROSIS 2023; 35:1-11. [PMID: 35738949 DOI: 10.1016/j.arteri.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMCs) undergo a phenotypic-switching process during the generation of unstable atheroma plaques. In this investigation, the potential implication of the tumor necrosis factor superfamily (TNFSF) ligands, in the gene expression signature associated with VSMC plasticity was studied. MATERIAL AND METHODS Human aortic (ha)VSMCs were obtained commercially and treated with the cytokine TNFSF14, also called LIGHT, the lymphotoxin alpha (LTα), the heterotrimer LTα1β2 or with vehicle for 72h. The effect of the different treatments on gene expression was analyzed by quantitative PCR and included the study of genes associated with myofibroblast-like cell function, osteochondrogenesis, pluripotency, lymphorganogenesis and macrophage-like cell function. RESULTS HaVSMCs displayed a change in myofibroblast-like cell genes which consisted in reduced COL1A1 and TGFB1 mRNA levels when treated with LTα or LIGHT and with augmented MMP9 expression levels when treated with LTα. LTα and LIGHT treatments also diminished the expression of genes associated with osteochondrogenesis and pluripotency SOX9, CKIT, and KLF4. By contrary, all the above genes were no affected by the treatment with the trimer LTα1β2. In addition, haVSMC treatment with LTα, LTα1β2 and LIGHT altered lymphorganogenic cytokine gene expression which consisted of augmented CCL20 and CCL21 mRNA levels by LTα and a reduction in the gene expression of CCL21 and CXCL13 by LIGHT and LTα1β2 respectively. Neither, LTα or LIGHT or LTα1β2 treatments affected the expression of macrophage-like cell markers in haVSMC. CONCLUSIONS Altogether, indicates that the TNFSF ligands through their interconnected network of signaling, are important in the preservation of VSMC identity against the acquisition of a genetic expression signature compatible with functional cellular plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Angela Vinué
- Institute of Health Research-INCLIVA, Valencia, Spain
| | - Sergio Martínez-Hervás
- Institute of Health Research-INCLIVA, Valencia, Spain; Endocrinology and Nutrition Department Clinic Hospital and Department of Medicine, University of Valencia, Valencia, Spain; CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Herminia González-Navarro
- Institute of Health Research-INCLIVA, Valencia, Spain; CIBER de Diabetes y Enfermedades Metabólicas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain; Biochemistry and Molecular Biology Department, Faculty of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
64
|
Bashore AC, Zhu LY, Reilly MP. A new era in understanding atherosclerotic plaques. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1127-1129. [PMID: 39196164 DOI: 10.1038/s44161-022-00187-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Alexander C Bashore
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA.
| | - Lucie Y Zhu
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY, USA
- The Irving Institute for Clinical and Translational Sciences, Columbia University, New York, NY, USA
| |
Collapse
|
65
|
Mokry M, Boltjes A, Slenders L, Bel-Bordes G, Cui K, Brouwer E, Mekke JM, Depuydt MA, Timmerman N, Waissi F, Verwer MC, Turner AW, Khan MD, Hodonsky CJ, Benavente ED, Hartman RJ, van den Dungen NAM, Lansu N, Nagyova E, Prange KH, Kovacic JC, Björkegren JL, Pavlos E, Andreakos E, Schunkert H, Owens GK, Monaco C, Finn AV, Virmani R, Leeper NJ, de Winther MP, Kuiper J, de Borst GJ, Stroes ES, Civelek M, de Kleijn DP, den Ruijter HM, Asselbergs FW, van der Laan SW, Miller CL, Pasterkamp G. Transcriptomic-based clustering of human atherosclerotic plaques identifies subgroups with different underlying biology and clinical presentation. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1140-1155. [PMID: 37920851 PMCID: PMC10621615 DOI: 10.1038/s44161-022-00171-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/20/2022] [Indexed: 11/04/2023]
Abstract
Histopathological studies have revealed key processes of atherosclerotic plaque thrombosis. However, the diversity and complexity of lesion types highlight the need for improved sub-phenotyping. Here we analyze the gene expression profiles of 654 advanced human carotid plaques. The unsupervised, transcriptome-driven clustering revealed five dominant plaque types. These plaque phenotypes were associated with clinical presentation and showed differences in cellular compositions. Validation in coronary segments showed that the molecular signature of these plaques was linked to coronary ischemia. One of the plaque types with the most severe clinical symptoms pointed to both inflammatory and fibrotic cell lineages. Further, we did a preliminary analysis of potential circulating biomarkers that mark the different plaques phenotypes. In conclusion, the definition of the plaque at risk for a thrombotic event can be fine-tuned by in-depth transcriptomic-based phenotyping. These differential plaque phenotypes prove clinically relevant for both carotid and coronary artery plaques and point to distinct underlying biology of symptomatic lesions.
Collapse
Affiliation(s)
- Michal Mokry
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Arjan Boltjes
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Gemma Bel-Bordes
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Kai Cui
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Eli Brouwer
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Joost M. Mekke
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Marie A.C. Depuydt
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Nathalie Timmerman
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Farahnaz Waissi
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Maarten C Verwer
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Mohammad Daud Khan
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Chani J. Hodonsky
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Ernest Diez Benavente
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Robin J.G. Hartman
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Noortje A M van den Dungen
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Nico Lansu
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Emilia Nagyova
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Koen H.M. Prange
- Amsterdam University Medical Centers – location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Jason C. Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, Australia; and St Vincent’s Clinical School, University of New South Wales, Australia
| | - Johan L.M. Björkegren
- Department of Medicine, Karolinska Institutet, Karolinska Universitetssjukhuset, Huddinge, Sweden
- Department of Genetics & Genomic Sciences, Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029-6574, USA
| | - Eleftherios Pavlos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK e.V.), Partner Site Munich Heart Alliance, Munich, Germany
| | - Gary K. Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford
| | | | | | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, CA
| | - Menno P.J. de Winther
- Amsterdam University Medical Centers – location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam, The Netherlands
| | - Johan Kuiper
- Leiden Academic Centre for Drug Research, Division of Biotherapeutics, Leiden University, Leiden, The Netherlands
| | - Gert J. de Borst
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Location AMC, Amsterdam, the Netherlands
| | - Mete Civelek
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | | | - Hester M. den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Folkert W. Asselbergs
- Department of Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Health Data Research UK and Institute of Health Informatics, University College London, London, United Kingdom
| | - Sander W. van der Laan
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| |
Collapse
|
66
|
The Role of Hydrogen Sulfide in Plaque Stability. Antioxidants (Basel) 2022; 11:antiox11122356. [PMID: 36552564 PMCID: PMC9774534 DOI: 10.3390/antiox11122356] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
Atherosclerosis is the greatest contributor to cardiovascular events and is involved in the majority of deaths worldwide. Plaque rapture or erosion precipitates life-threatening thrombi, resulting in the obstruction blood flow to the heart (acute coronary syndrome), brain (ischemic stroke) or low extremities (peripheral vascular diseases). Among these events, major causation dues to the plaque rupture. Although the initiation, procession, and precise time of controlling plaque rupture are unclear, foam cell formation and apoptosis, cell death, extracellular matrix components, protease expression and activity, local inflammation, intraplaque hemorrhage, and calcification contribute to the plaque instability. These alterations tightly associate with the function regulation of intraplaque various cell populations. Hydrogen sulfide (H2S) is gasotransmitter derived from methionine metabolism and exerts a protective role in the genesis of atherosclerosis. Recent progress also showed H2S mediated the plaque stability. In this review, we discuss the progress of endogenous H2S modulation on functions of vascular smooth muscle cells, monocytes/macrophages, and T cells, and the molecular mechanism in plaque stability.
Collapse
|
67
|
Li Q, Wang M, Zhang S, Jin M, Chen R, Luo Y, Sun X. Single-cell RNA sequencing in atherosclerosis: Mechanism and precision medicine. Front Pharmacol 2022; 13:977490. [PMID: 36267275 PMCID: PMC9576927 DOI: 10.3389/fphar.2022.977490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis is the pathological basis of various vascular diseases, including those with high mortality, such as myocardial infarction and stroke. However, its pathogenesis is complex and has not been fully elucidated yet. Over the past few years, single-cell RNA sequencing (scRNA-seq) has been developed and widely used in many biological fields to reveal biological mechanisms at the cellular level and solve the problems of cellular heterogeneity that cannot be solved using bulk RNA sequencing. In this review, we briefly summarize the existing scRNA-seq technologies and focus on their application in atherosclerosis research to provide insights into the occurrence, development and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Qiaoyu Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Mengchen Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Shuxia Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Rongchang Chen
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
- *Correspondence: Yun Luo, ; Xiaobo Sun,
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- NMPA Key Laboratory for Research and Evaluation of Pharmacovigilance, Beijing, China
- *Correspondence: Yun Luo, ; Xiaobo Sun,
| |
Collapse
|
68
|
Gole S, Tkachenko S, Masannat T, Baylis RA, Cherepanova OA. Endothelial-to-Mesenchymal Transition in Atherosclerosis: Friend or Foe? Cells 2022; 11:cells11192946. [PMID: 36230908 PMCID: PMC9563961 DOI: 10.3390/cells11192946] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
Despite many decades of research, complications of atherosclerosis resulting from the rupture or erosion of unstable plaques remain the leading cause of death worldwide. Advances in cellular lineage tracing techniques have allowed researchers to begin investigating the role of individual cell types in the key processes regulating plaque stability, including maintenance of the fibrous cap, a protective collagen-rich structure that underlies the endothelium. This structure was previously thought to be entirely derived from smooth muscle cells (SMC), which migrated from the vessel wall. However, recent lineage tracing studies have identified endothelial cells (EC) as an essential component of this protective barrier through an endothelial-to-mesenchymal transition (EndoMT), a process that has previously been implicated in pulmonary, cardiac, and kidney fibrosis. Although the presence of EndoMT in atherosclerotic plaques has been shown by several laboratories using EC-lineage tracing mouse models, whether EndoMT is detrimental (i.e., worsening disease progression) or beneficial (i.e., an athero-protective response that prevents plaque instability) remains uncertain as there are data to support both possibilities, which will be further discussed in this review.
Collapse
Affiliation(s)
- Sarin Gole
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NB5, Cleveland, OH 44195, USA
| | - Svyatoslav Tkachenko
- Genetics and Genome Sciences, Case Western Reserve University, 2109 Adelbert, RD, BRB, Cleveland, OH 44106, USA
| | - Tarek Masannat
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NB5, Cleveland, OH 44195, USA
| | - Richard A. Baylis
- Department of Medicine, Massachusetts General Hospital, 55 Fruit St Gray 730, Boston, MA 02114, USA
| | - Olga A. Cherepanova
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, NB5, Cleveland, OH 44195, USA
- Correspondence: ; Tel.: +1-216-445-7491
| |
Collapse
|
69
|
Kawai K, Vozenilek AE, Kawakami R, Sato Y, Ghosh SKB, Virmani R, Finn AV. Understanding the role of alternative macrophage phenotypes in human atherosclerosis. Expert Rev Cardiovasc Ther 2022; 20:689-705. [PMID: 35942866 DOI: 10.1080/14779072.2022.2111301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
INTRODUCTION Atherosclerosis-based ischemic heart disease is still the primary cause of death throughout the world. Over the past decades there has been no significant changes in the therapeutic approaches to atherosclerosis, which are mainly based on lipid lowering therapies and management of comorbid conditions such as diabetes and hypertension. The involvement of macrophages in atherosclerosis has been recognized for decades. More recently, a more detailed and sophisticated understanding of their various phenotypes and roles in the atherosclerotic process has been recognized. This new data is revealing how specific subtypes of macrophage-induced inflammation may have distinct effects on atherosclerosis progression and may provide new approaches for treatment, based upon targeting of specific macrophage subtypes. AREAS COVERED We will comprehensively review the spectrum of macrophage phenotypes and how they contribute to atherosclerotic plaque development and progression. EXPERT OPINION Various signals derived from atherosclerotic lesions drive macrophages into complex subsets with different gene expression profiles, phenotypes, and functions, not all of which are understood. Macrophage phenotypes include those that enhance, heal, and regress the atherosclerotic lesions though various mechanisms. Targeting of specific macrophage phenotypes may provide a promising and novel approach to prevent atherosclerosis progression.
Collapse
Affiliation(s)
- Kenji Kawai
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aimee E Vozenilek
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Rika Kawakami
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Yu Sato
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | | | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA
| | - Aloke V Finn
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD, USA.,University of Maryland, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
70
|
Li L, Wang M, Ma Q, Ye J, Sun G. Role of glycolysis in the development of atherosclerosis. Am J Physiol Cell Physiol 2022; 323:C617-C629. [DOI: 10.1152/ajpcell.00218.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a chronic inflammatory vascular disease associated with atherosclerotic plaques and endothelial dysfunction, inflammation, and plaque formation. Glycolysis is a conservative and rigorous biological process that decomposes glucose into pyruvate. Its function is to provide the body with energy and intermediate products required for life activities. However, abnormalities in glycolytic flux during the progression of atherosclerosis accelerate disease progression. Here, we review the role of glycolysis in the development of atherosclerosis to provide new ideas for developing novel anti-atherosclerosis strategies.
Collapse
Affiliation(s)
- Lanfang Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Qiuxiao Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingxue Ye
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
71
|
Elishaev M, Hodonsky CJ, Ghosh SKB, Finn AV, von Scheidt M, Wang Y. Opportunities and Challenges in Understanding Atherosclerosis by Human Biospecimen Studies. Front Cardiovasc Med 2022; 9:948492. [PMID: 35872917 PMCID: PMC9300954 DOI: 10.3389/fcvm.2022.948492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over the last few years, new high-throughput biotechnologies and bioinformatic methods are revolutionizing our way of deep profiling tissue specimens at the molecular levels. These recent innovations provide opportunities to advance our understanding of atherosclerosis using human lesions aborted during autopsies and cardiac surgeries. Studies on human lesions have been focusing on understanding the relationship between molecules in the lesions with tissue morphology, genetic risk of atherosclerosis, and future adverse cardiovascular events. This review will highlight ways to utilize human atherosclerotic lesions in translational research by work from large cardiovascular biobanks to tissue registries. We will also discuss the opportunities and challenges of working with human atherosclerotic lesions in the era of next-generation sequencing.
Collapse
Affiliation(s)
- Maria Elishaev
- Department of Pathology and Laboratory Medicine, Center for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| | - Chani J. Hodonsky
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| | | | - Aloke V. Finn
- Cardiovascular Pathology Institute, Gaithersburg, MD, United States
| | - Moritz von Scheidt
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, Munich, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung, Partner Site Munich Heart Alliance, Munich, Germany
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, Center for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
72
|
Qin W, Li F, Jia L, Wang Q, Li Y, Wei Y, Li Y, Jin H, Jia J. Phosphorylated Tau 181 Serum Levels Predict Alzheimer’s Disease in the Preclinical Stage. Front Aging Neurosci 2022; 14:900773. [PMID: 35769604 PMCID: PMC9234327 DOI: 10.3389/fnagi.2022.900773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background There is an urgent need for cost-effective, easy-to-measure biomarkers to identify subjects who will develop Alzheimer’s disease (AD), especially at the pre-symptomatic stage. This stage can be determined in autosomal dominant AD (ADAD) which offers the opportunity to observe the dynamic biomarker changes during the life-course of AD stages. This study aimed to investigate serum biomarkers during different AD stages and potential novel protein biomarkers of presymptomatic AD. Methods In the first stage, 32 individuals [20 mutation carriers including 10 with AD, and 10 with mild cognitive impairment (MCI), and 12 healthy controls] from ADAD families were analyzed. All subjects underwent a complete clinical evaluation and a comprehensive neuropsychological battery. Serum samples were collected from all subjects, and antibody arrays were used to analyze 170 proteins in these samples. The most promising biomarkers were identified during this screening and were then measured in serum samples of 12 subjects with pre-MCI and 20 controls. Results The serum levels of 13 proteins were significantly different in patients with AD or MCI compared to controls. Of the 13 proteins, cathepsin D, immunoglobulin E, epidermal growth factor receptor (EGFR), matrix metalloproteinase-9 (MMP-9), von Willebrand factor (vWF), haptoglobin, and phosphorylated Tau-181 (p-Tau181) correlated with all cognitive measures (R2 = −0.69–0.76). The areas under the receiver operating characteristic curve of these seven proteins were 0.71–0.93 for the classification of AD and 0.57–0.95 for the classification of MCI. Higher levels of p-Tau181 were found in the serum of pre-MCI subjects than in the serum of controls. The p-Tau181 serum level might detect AD before symptoms occur (area under the curve 0.85, sensitivity 75%, specificity 81.67%). Conclusions A total of 13 serum proteins showed significant differences between subjects with AD and MCI and healthy controls. The p-Tau181 serum level might be a broadly available and cost-effective biomarker to identify individuals with preclinical AD and assess the severity of AD.
Collapse
Affiliation(s)
- Wei Qin
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Fangyu Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Longfei Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qi Wang
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Ying Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yiping Wei
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yan Li
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Hongmei Jin
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Jianping Jia
- Innovation Center for Neurological Disorders and Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Beijing Key Laboratory of Geriatric Cognitive Disorders, Capital Medical University, Beijing, China
- Clinical Center for Neurodegenerative Disease and Memory Impairment, Capital Medical University, Beijing, China
- Center of Alzheimer’s Disease, Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
- *Correspondence: Jianping Jia
| |
Collapse
|
73
|
Björkegren JLM, Lusis AJ. Atherosclerosis: Recent developments. Cell 2022; 185:1630-1645. [PMID: 35504280 PMCID: PMC9119695 DOI: 10.1016/j.cell.2022.04.004] [Citation(s) in RCA: 385] [Impact Index Per Article: 192.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022]
Abstract
Atherosclerosis is an inflammatory disease of the large arteries that is the major cause of cardiovascular disease (CVD) and stroke. Here, we review the current understanding of the molecular, cellular, genetic, and environmental contributions to atherosclerosis, from both individual pathway and systems perspectives. We place an emphasis on recent developments, some of which have yielded unexpected biology, including previously unknown heterogeneity of inflammatory and smooth muscle cells in atherosclerotic lesions, roles for senescence and clonal hematopoiesis, and links to the gut microbiome.
Collapse
Affiliation(s)
- Johan L M Björkegren
- Department of Genetics and Genomic Sciences, Division of Cardiology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, Department of Microbiology, Immunology and Molecular Genetics, Department of Human Genetics, A2-237 Center for the Health Sciences, University of California, Los Angeles, Los Angeles, CA USA.
| |
Collapse
|
74
|
Howe KL, Cybulsky M, Fish JE. The Endothelium as a Hub for Cellular Communication in Atherogenesis: Is There Directionality to the Message? Front Cardiovasc Med 2022; 9:888390. [PMID: 35498030 PMCID: PMC9051343 DOI: 10.3389/fcvm.2022.888390] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells line every blood vessel and thereby serve as an interface between the blood and the vessel wall. They have critical functions for maintaining homeostasis and orchestrating vascular pathogenesis. Atherosclerosis is a chronic disease where cholesterol and inflammatory cells accumulate in the artery wall below the endothelial layer and ultimately form plaques that can either progress to occlude the lumen or rupture with thromboembolic consequences - common outcomes being myocardial infarction and stroke. Cellular communication lies at the core of this process. In this review, we discuss traditional (e.g., cytokines, chemokines, nitric oxide) and novel (e.g., extracellular vesicles) modes of endothelial communication with other endothelial cells as well as circulating and vessel wall cells, including monocytes, macrophages, neutrophils, vascular smooth muscle cells and other immune cells, in the context of atherosclerosis. More recently, the growing appreciation of endothelial cell plasticity during atherogenesis suggests that communication strategies are not static. Here, emerging data on transcriptomics in cells during the development of atherosclerosis are considered in the context of how this might inform altered cell-cell communication. Given the unique position of the endothelium as a boundary layer that is activated in regions overlying vascular inflammation and atherosclerotic plaque, there is a potential to exploit the unique features of this group of cells to deliver therapeutics that target the cellular crosstalk at the core of atherosclerotic disease. Data are discussed supporting this concept, as well as inherent pitfalls. Finally, we briefly review the literature for other regions of the body (e.g., gut epithelium) where cells similarly exist as a boundary layer but provide discrete messages to each compartment to govern homeostasis and disease. In this light, the potential for endothelial cells to communicate in a directional manner is explored, along with the implications of this concept - from fundamental experimental design to biomarker potential and therapeutic targets.
Collapse
Affiliation(s)
- Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Myron Cybulsky
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
75
|
Cheng P, Wirka RC, Kim JB, Kim HJ, Nguyen T, Kundu R, Zhao Q, Sharma D, Pedroza A, Nagao M, Iyer D, Fischbein MP, Quertermous T. Smad3 regulates smooth muscle cell fate and mediates adverse remodeling and calcification of the atherosclerotic plaque. NATURE CARDIOVASCULAR RESEARCH 2022; 1:322-333. [PMID: 36246779 PMCID: PMC9560061 DOI: 10.1038/s44161-022-00042-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 03/01/2022] [Indexed: 04/20/2023]
Abstract
Atherosclerotic plaques consist mostly of smooth muscle cells (SMC), and genes that influence SMC phenotype can modulate coronary artery disease (CAD) risk. Allelic variation at 15q22.33 has been identified by genome-wide association studies to modify the risk of CAD and is associated with the expression of SMAD3 in SMC. However, the mechanism by which this gene modifies CAD risk remains poorly understood. Here we show that SMC-specific deletion of Smad3 in a murine atherosclerosis model resulted in greater plaque burden, more outward remodelling and increased vascular calcification. Single-cell transcriptomic analyses revealed that loss of Smad3 altered SMC transition cell state toward two fates: a SMC phenotype that governs both vascular remodelling and recruitment of inflammatory cells, as well as a chondromyocyte fate. Together, the findings reveal that Smad3 expression in SMC inhibits the emergence of specific SMC phenotypic transition cells that mediate adverse plaque features, including outward remodelling, monocyte recruitment, and vascular calcification.
Collapse
Affiliation(s)
- Paul Cheng
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Robert C. Wirka
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Hyun-Jung Kim
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Trieu Nguyen
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Ramendra Kundu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Quanyi Zhao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Disha Sharma
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Albert Pedroza
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Manabu Nagao
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Dharini Iyer
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Michael P. Fischbein
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305
| | - Thomas Quertermous
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
76
|
Metabolism in atherosclerotic plaques: immunoregulatory mechanisms in the arterial wall. Clin Sci (Lond) 2022; 136:435-454. [PMID: 35348183 PMCID: PMC8965849 DOI: 10.1042/cs20201293] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/02/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023]
Abstract
Over the last decade, there has been a growing interest to understand the link between metabolism and the immune response in the context of metabolic diseases but also beyond, giving then birth to a new field of research. Termed 'immunometabolism', this interdisciplinary field explores paradigms of both immunology and metabolism to provided unique insights into different disease pathogenic processes, and the identification of new potential therapeutic targets. Similar to other inflammatory conditions, the atherosclerotic inflammatory process in the artery has been associated with a local dysregulated metabolic response. Thus, recent studies show that metabolites are more than just fuels in their metabolic pathways, and they can act as modulators of vascular inflammation and atherosclerosis. In this review article, we describe the most common immunometabolic pathways characterised in innate and adaptive immune cells, and discuss how macrophages' and T cells' metabolism may influence phenotypic changes in the plaque. Moreover, we discuss the potential of targeting immunometabolism to prevent and treat cardiovascular diseases (CVDs).
Collapse
|
77
|
Alvandi Z, Nagata Y, Passos LSA, Hashemi Gheinani A, Guerrero JL, Wylie‐Sears J, Romero DC, Morris BA, Sullivan SM, Yaghoubian KM, Alvandi A, Adam RM, Aikawa E, Levine RA, Bischoff J. Wnt Site Signaling Inhibitor Secreted Frizzled‐Related Protein 3 Protects Mitral Valve Endothelium From Myocardial Infarction–Induced Endothelial‐to‐Mesenchymal Transition. J Am Heart Assoc 2022; 11:e023695. [PMID: 35348006 PMCID: PMC9075477 DOI: 10.1161/jaha.121.023695] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background
The onset and mechanisms of endothelial‐to‐mesenchymal transition (EndMT) in mitral valve (MV) leaflets following myocardial infarction (MI) are unknown, yet these events are closely linked to stiffening of leaflets and development of ischemic mitral regurgitation. We investigated whether circulating molecules present in plasma within days after MI incite EndMT in MV leaflets.
Methods and Results
We examined the onset of EndMT in MV leaflets from 9 sheep with inferior MI, 8 with sham surgery, and 6 naïve controls. Ovine MVs 8 to 10 days after inferior MI displayed EndMT, shown by increased vascular endothelial cadherin/α‐smooth muscle actin–positive cells. The effect of plasma on EndMT in MV endothelial cells (VECs) was assessed by quantitative polymerase chain reaction, migration assays, and immunofluorescence. In vitro, post‐MI plasma induced EndMT marker expression and enhanced migration of mitral VECs; sham plasma did not. Analysis of sham versus post‐MI plasma revealed a significant drop in the Wnt signaling antagonist sFRP3 (secreted frizzled‐related protein 3) in post‐MI plasma. Addition of recombinant sFRP3 to post‐MI plasma reversed its EndMT‐inducing effect on mitral VECs. RNA‐sequencing analysis of mitral VECs exposed to post‐MI plasma showed upregulated FOXM1 (forkhead box M1). Blocking FOXM1 reduced EndMT transcripts in mitral VECs treated with post‐MI plasma. Finally, FOXM1 induced by post‐MI plasma was downregulated by sFRP3.
Conclusions
Reduced sFRP3 in post‐MI plasma facilitates EndMT in mitral VECs by increasing the transcription factor FOXM1. Restoring sFRP3 levels or inhibiting FOXM1 soon after MI may provide a novel strategy to modulate EndMT in the MV to prevent ischemic mitral regurgitation and heart failure.
Collapse
Affiliation(s)
- Zahra Alvandi
- Vascular Biology Program Boston Children’s Hospital Boston MA
- Department of Surgery Harvard Medical School Boston MA
| | - Yasufumi Nagata
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | | | - Ali Hashemi Gheinani
- Department of Surgery Harvard Medical School Boston MA
- Broad Institute of MIT and Harvard Cambridge MA
- Department of Urology Boston Children’s Hospital Boston MA
| | - J. Luis Guerrero
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | | | - Dayana Carolina Romero
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | - Brittan A. Morris
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | - Suzanne M. Sullivan
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | - Koushiar M. Yaghoubian
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | - Amirhossein Alvandi
- Department of Mathematics and Statistics University of Massachusetts Amherst MA
| | - Rosalyn M. Adam
- Department of Surgery Harvard Medical School Boston MA
- Department of Urology Boston Children’s Hospital Boston MA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology Brigham and Women’s Hospital Harvard Medical School Boston MA
- Center for Interdisciplinary Cardiovascular Sciences Cardiovascular MedicineBrigham and Women’s HospitalHarvard Medical School Boston MA
| | - Robert A. Levine
- Cardiac Ultrasound Laboratory Massachusetts General HospitalHarvard Medical School Boston MA
| | - Joyce Bischoff
- Vascular Biology Program Boston Children’s Hospital Boston MA
- Department of Surgery Harvard Medical School Boston MA
| |
Collapse
|
78
|
McQueen LW, Ladak SS, Abbasciano R, George SJ, Suleiman MS, Angelini GD, Murphy GJ, Zakkar M. Next-Generation and Single-Cell Sequencing Approaches to Study Atherosclerosis and Vascular Inflammation Pathophysiology: A Systematic Review. Front Cardiovasc Med 2022; 9:849675. [PMID: 35419441 PMCID: PMC8996078 DOI: 10.3389/fcvm.2022.849675] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Background and Aims Atherosclerosis is a chronic inflammatory disease that remains the leading cause of morbidity and mortality worldwide. Despite decades of research into the development and progression of this disease, current management and treatment approaches remain unsatisfactory and further studies are required to understand the exact pathophysiology. This review aims to provide a comprehensive assessment of currently published data utilizing single-cell and next-generation sequencing techniques to identify key cellular and molecular contributions to atherosclerosis and vascular inflammation. Methods Electronic searches of Cochrane Central Register of Controlled Trials, MEDLINE, and EMBASE databases were undertaken from inception until February 2022. A narrative synthesis of all included studies was performed for all included studies. Quality assessment and risk of bias analysis was evaluated using the ARRIVE and SYRCLE checklist tools. Results Thirty-four studies were eligible for narrative synthesis, with 16 articles utilizing single-cell exclusively, 10 utilizing next-generation sequencing and 8 using a combination of these approaches. Studies investigated numerous targets, ranging from exploratory tissue and plaque analysis, cell phenotype investigation and physiological/hemodynamic contributions to disease progression at both the single-cell and whole genome level. A significant area of focus was placed on smooth muscle cell, macrophage, and stem/progenitor contributions to disease, with little focus placed on contributions of other cell types including lymphocytes and endothelial cells. A significant level of heterogeneity exists in the outcomes from single-cell sequencing of similar samples, leading to inter-sample and inter-study variation. Conclusions Single-cell and next-generation sequencing methodologies offer novel means of elucidating atherosclerosis with significantly higher resolution than previous methodologies. These approaches also show significant potential for translatability into other vascular disease states, by facilitating cell-specific gene expression profiles between disease states. Implementation of these technologies may offer novel approaches to understanding the disease pathophysiology and improving disease prevention, management, and treatment.Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021229960, identifier: CRD42021229960.
Collapse
Affiliation(s)
- Liam W. McQueen
- Department of Cardiovascular Sciences, Clinical Science Wing, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| | - Shameem S. Ladak
- Department of Cardiovascular Sciences, Clinical Science Wing, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| | - Riccardo Abbasciano
- Department of Cardiovascular Sciences, Clinical Science Wing, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| | - Sarah J. George
- Bristol Heart Institute and Translational Biomedical Research Centre, Bristol Medical School, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - M-Saadeh Suleiman
- Bristol Heart Institute and Translational Biomedical Research Centre, Bristol Medical School, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - Gianni D. Angelini
- Bristol Heart Institute and Translational Biomedical Research Centre, Bristol Medical School, Bristol Royal Infirmary, University of Bristol, Bristol, United Kingdom
| | - Gavin J. Murphy
- Department of Cardiovascular Sciences, Clinical Science Wing, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| | - Mustafa Zakkar
- Department of Cardiovascular Sciences, Clinical Science Wing, Glenfield Hospital, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
79
|
Shin J, Tkachenko S, Chaklader M, Pletz C, Singh K, Bulut GB, Han YM, Mitchell K, Baylis RA, Kuzmin AA, Hu B, Lathia JD, Stenina-Adognravi O, Podrez E, Byzova TV, Owens GK, Cherepanova OA. Endothelial OCT4 is atheroprotective by preventing metabolic and phenotypic dysfunction. Cardiovasc Res 2022; 118:2458-2477. [PMID: 35325071 PMCID: PMC9890633 DOI: 10.1093/cvr/cvac036] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 02/04/2023] Open
Abstract
AIMS Until recently, the pluripotency factor Octamer (ATGCAAAT)-binding transcriptional factor 4 (OCT4) was believed to be dispensable in adult somatic cells. However, our recent studies provided clear evidence that OCT4 has a critical atheroprotective role in smooth muscle cells. Here, we asked if OCT4 might play a functional role in regulating endothelial cell (EC) phenotypic modulations in atherosclerosis. METHODS AND RESULTS Specifically, we show that EC-specific Oct4 knockout resulted in increased lipid, LGALS3+ cell accumulation, and altered plaque characteristics consistent with decreased plaque stability. A combination of single-cell RNA sequencing and EC-lineage-tracing studies revealed increased EC activation, endothelial-to-mesenchymal transitions, plaque neovascularization, and mitochondrial dysfunction in the absence of OCT4. Furthermore, we show that the adenosine triphosphate (ATP) transporter, ATP-binding cassette (ABC) transporter G2 (ABCG2), is a direct target of OCT4 in EC and establish for the first time that the OCT4/ABCG2 axis maintains EC metabolic homeostasis by regulating intracellular heme accumulation and related reactive oxygen species production, which, in turn, contributes to atherogenesis. CONCLUSIONS These results provide the first direct evidence that OCT4 has a protective metabolic function in EC and identifies vascular OCT4 and its signalling axis as a potential target for novel therapeutics.
Collapse
Affiliation(s)
| | | | | | - Connor Pletz
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kanwardeep Singh
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gamze B Bulut
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Young min Han
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, USA
| | - Kelly Mitchell
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Richard A Baylis
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Andrey A Kuzmin
- Russian Academy of Sciences, Institute of Cytology, St Petersburg, Russian Federation
| | - Bo Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga Stenina-Adognravi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eugene Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tatiana V Byzova
- Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA,Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | | |
Collapse
|
80
|
Platelets, a Key Cell in Inflammation and Atherosclerosis Progression. Cells 2022; 11:cells11061014. [PMID: 35326465 PMCID: PMC8947573 DOI: 10.3390/cells11061014] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 12/21/2022] Open
Abstract
Platelets play important roles in thrombosis-dependent obstructive cardiovascular diseases. In addition, it has now become evident that platelets also participate in the earliest stages of atherosclerosis, including the genesis of the atherosclerotic lesion. Moreover, while the link between platelet activity and hemostasis has been well established, the role of platelets as modulators of inflammation has only recently been recognized. Thus, through their secretory activities, platelets can chemically attract a diverse repertoire of cells to inflammatory foci. Although monocytes and lymphocytes act as key cells in the progression of an inflammatory event and play a central role in plaque formation and progression, there is also evidence that platelets can traverse the endothelium, and therefore be a direct mediator in the progression of atherosclerotic plaque. This review provides an overview of platelet interactions and regulation in atherosclerosis.
Collapse
|
81
|
Misra A, Rehan R, Lin A, Patel S, Fisher EA. Emerging Concepts of Vascular Cell Clonal Expansion in Atherosclerosis. Arterioscler Thromb Vasc Biol 2022; 42:e74-e84. [PMID: 35109671 PMCID: PMC8988894 DOI: 10.1161/atvbaha.121.316093] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clonal expansion is a process that can drive pathogenesis in human diseases, with atherosclerosis being a prominent example. Despite advances in understanding the etiology of atherosclerosis, clonality studies of vascular cells remain in an early stage. Recently, several paradigm-shifting preclinical studies have identified clonal expansion of progenitor cells in the vasculature in response to atherosclerosis. This review provides an overview of cell clonality in atherosclerotic progression, focusing particularly on smooth muscle cells and macrophages. We discuss key findings from the latest research that give insight into the mechanisms by which clonal expansion of vascular cells contributes to disease pathology. The further probing of these mechanisms will provide innovative directions for future progress in the understanding and therapy of atherosclerosis and its associated cardiovascular diseases.
Collapse
Affiliation(s)
- Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rajan Rehan
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia,Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Alexander Lin
- Heart Research Institute, Sydney, NSW 2042, Australia,School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia,Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia,Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Edward A Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, NY, USA,Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
82
|
Cheng P, Wirka RC, Clarke LS, Zhao Q, Kundu R, Nguyen T, Nair S, Sharma D, Kim HJ, Shi H, Assimes T, Kim JB, Kundaje A, Quertermous T. ZEB2 Shapes the Epigenetic Landscape of Atherosclerosis. Circulation 2022; 145:469-485. [PMID: 34990206 PMCID: PMC8896308 DOI: 10.1161/circulationaha.121.057789] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND Smooth muscle cells (SMCs) transition into a number of different phenotypes during atherosclerosis, including those that resemble fibroblasts and chondrocytes, and make up the majority of cells in the atherosclerotic plaque. To better understand the epigenetic and transcriptional mechanisms that mediate these cell state changes, and how they relate to risk for coronary artery disease (CAD), we have investigated the causality and function of transcription factors at genome-wide associated loci. METHODS We used CRISPR-Cas 9 genome and epigenome editing to identify the causal gene and cells for a complex CAD genome-wide association study signal at 2q22.3. Single-cell epigenetic and transcriptomic profiling in murine models and human coronary artery smooth muscle cells were used to understand the cellular and molecular mechanism by which this CAD risk gene exerts its function. RESULTS CRISPR-Cas 9 genome and epigenome editing showed that the complex CAD genetic signals within a genomic region at 2q22.3 lie within smooth muscle long-distance enhancers for ZEB2, a transcription factor extensively studied in the context of epithelial mesenchymal transition in development of cancer. Zeb2 regulates SMC phenotypic transition through chromatin remodeling that obviates accessibility and disrupts both Notch and transforming growth factor β signaling, thus altering the epigenetic trajectory of SMC transitions. SMC-specific loss of Zeb2 resulted in an inability of transitioning SMCs to turn off contractile programing and take on a fibroblast-like phenotype, but accelerated the formation of chondromyocytes, mirroring features of high-risk atherosclerotic plaques in human coronary arteries. CONCLUSIONS These studies identify ZEB2 as a new CAD genome-wide association study gene that affects features of plaque vulnerability through direct effects on the epigenome, providing a new therapeutic approach to target vascular disease.
Collapse
Affiliation(s)
- Paul Cheng
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Robert C. Wirka
- Division of Cardiology, Departments of Medicine and Cell Biology and Physiology, McAllister Heart Institute, University of North Carolina, Chapel Hill, NC
| | - Lee Shoa Clarke
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Quanyi Zhao
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Ramendra Kundu
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Trieu Nguyen
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Surag Nair
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Disha Sharma
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Hyun-jung Kim
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Huitong Shi
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Themistocles Assimes
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| | - Anshul Kundaje
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305
| | - Thomas Quertermous
- Division of Cardiovascular Medicine and the Cardiovascular Institute, Stanford, CA
| |
Collapse
|
83
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
84
|
Hartmann F, Gorski DJ, Newman AAC, Homann S, Petz A, Owsiany KM, Serbulea V, Zhou YQ, Deaton RA, Bendeck M, Owens GK, Fischer JW. SMC-Derived Hyaluronan Modulates Vascular SMC Phenotype in Murine Atherosclerosis. Circ Res 2021; 129:992-1005. [PMID: 34615369 DOI: 10.1161/circresaha.120.318479] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Felicia Hartmann
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany (F.H., D.J.G., S.H., A.P., J.W.F.)
| | - Daniel J Gorski
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany (F.H., D.J.G., S.H., A.P., J.W.F.)
| | - Alexandra A C Newman
- Robert M. Berne Cardiovascular Research Center (A.A.C.N., K.M.O., V.S., R.A.D., G.K.O), University of Virginia-School of Medicine, Charlottesville.,Department of Biochemistry and Molecular Genetics (A.A.C.N., K.M.O.), University of Virginia-School of Medicine, Charlottesville.,Cardiovascular Research Center in the Department of Medicine, New York University (A.A.C.N.)
| | - Susanne Homann
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany (F.H., D.J.G., S.H., A.P., J.W.F.)
| | - Anne Petz
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany (F.H., D.J.G., S.H., A.P., J.W.F.)
| | - Katherine M Owsiany
- Robert M. Berne Cardiovascular Research Center (A.A.C.N., K.M.O., V.S., R.A.D., G.K.O), University of Virginia-School of Medicine, Charlottesville.,Department of Biochemistry and Molecular Genetics (A.A.C.N., K.M.O.), University of Virginia-School of Medicine, Charlottesville
| | - Vlad Serbulea
- Robert M. Berne Cardiovascular Research Center (A.A.C.N., K.M.O., V.S., R.A.D., G.K.O), University of Virginia-School of Medicine, Charlottesville
| | - Yu-Qing Zhou
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (Y.Q.-Z., M.B.)
| | - Rebecca A Deaton
- Robert M. Berne Cardiovascular Research Center (A.A.C.N., K.M.O., V.S., R.A.D., G.K.O), University of Virginia-School of Medicine, Charlottesville
| | - Michelle Bendeck
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada (Y.Q.-Z., M.B.)
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center (A.A.C.N., K.M.O., V.S., R.A.D., G.K.O), University of Virginia-School of Medicine, Charlottesville
| | - Jens W Fischer
- Institute of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Germany (F.H., D.J.G., S.H., A.P., J.W.F.)
| |
Collapse
|
85
|
Jeong K, Murphy JM, Kim JH, Campbell PM, Park H, Rodriguez Y, Choi C, Kim JS, Park S, Kim HJ, Scammell JG, Weber DS, Honkanen RE, Schlaepfer DD, Ahn EYE, Lim STS. FAK Activation Promotes SMC Dedifferentiation via Increased DNA Methylation in Contractile Genes. Circ Res 2021; 129:e215-e233. [PMID: 34702049 DOI: 10.1161/circresaha.121.319066] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale: Vascular smooth muscle cells (SMCs) exhibit remarkable plasticity and can undergo dedifferentiation upon pathological stimuli associated with disease and interventions. Objective: Although epigenetic changes are critical in SMC phenotype switching, a fundamental regulator that governs the epigenetic machineries regulating the fate of SMC phenotype has not been elucidated. Methods and Results: Using SMCs, mouse models, and human atherosclerosis specimens, we found that focal adhesion kinase (FAK) activation elicits SMC dedifferentiation by stabilizing DNA methyltransferase 3A (DNMT3A). FAK in SMCs is activated in the cytoplasm upon serum stimulation in vitro or vessel injury and active FAK prevents DNMT3A from nuclear FAK-mediated degradation. However, pharmacological or genetic FAK catalytic inhibition forced FAK nuclear localization, which reduced DNMT3A protein via enhanced ubiquitination and proteasomal degradation. Reduced DNMT3A protein led to DNA hypomethylation in contractile gene promoters, which increased SMC contractile protein expression. RNA sequencing identified SMC contractile genes as a foremost upregulated group by FAK inhibition from injured femoral artery samples compared to vehicle group. DNMT3A knockdown in injured arteries reduced DNA methylation and enhanced contractile gene expression supports the notion that nuclear FAK-mediated DNMT3A degradation via E3 ligase TRAF6 drives differentiation of SMCs. Furthermore, we observed that SMCs of human atherosclerotic lesions exhibited decreased nuclear FAK, which was associated with increased DNMT3A levels and decreased contractile gene expression. Conclusions: This study reveals that nuclear FAK induced by FAK catalytic inhibition specifically suppresses DNMT3A expression in injured vessels resulting in maintaining SMC differentiation by promoting the contractile gene expression. Thus, FAK inhibitors may provide a new treatment option to block SMC phenotypic switching during vascular remodeling and atherosclerosis.
Collapse
Affiliation(s)
- Kyuho Jeong
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | - James M Murphy
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | - Jung-Hyun Kim
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | | | - Hyeonsoo Park
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, KOREA, REPUBLIC OF
| | - Yelitza Rodriguez
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | - Chungsik Choi
- Physiology, University of South Alabama College of Medicine, UNITED STATES
| | - Jun-Sub Kim
- Biotechnology, Korea National University of Transportation, KOREA, REPUBLIC OF
| | - Sangwon Park
- Pharmacology, Gyeongsang National University, KOREA, REPUBLIC OF
| | - Hyun Joon Kim
- Anatomy and Convergence Medical Sciences, Gyeongsang National University
| | - Jonathan G Scammell
- Comparative Medicine, University of South Alabama College of Medicine, UNITED STATES
| | - David S Weber
- Physiology and Cell Biology, University of South Alabama College of Medicine, UNITED STATES
| | - Richard E Honkanen
- Biochemistry and Molecualr Biology, University of South Alabama College of Medicine, UNITED STATES
| | - David D Schlaepfer
- Obstetrics, Gynecology, and Reproductive Medicine, University of California, San Diego Moores Cancer Center, UNITED STATES
| | | | - Ssang-Taek Steve Lim
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| |
Collapse
|
86
|
Buono MF, Slenders L, Wesseling M, Hartman RJG, Monaco C, den Ruijter HM, Pasterkamp G, Mokry M. The changing landscape of the vulnerable plaque: a call for fine-tuning of preclinical models. Vascul Pharmacol 2021; 141:106924. [PMID: 34607015 DOI: 10.1016/j.vph.2021.106924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/08/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022]
Abstract
For decades, the pathological definition of the vulnerable plaque led to invaluable insights into the mechanisms that underlie myocardial infarction and stroke. Beyond plaque rupture, other mechanisms, such as erosion, may elicit thrombotic events underlining the complexity and diversity of the atherosclerotic disease. Novel insights, based on single-cell transcriptomics and other "omics" methods, provide tremendous opportunities in the ongoing search for cell-specific determinants that will fine-tune the description of the thrombosis prone lesion. It coincides with an increasing awareness that knowledge on lesion characteristics, cell plasticity and clinical presentation of ischemic cardiovascular events have shifted over the past decades. This shift correlates with an observed changes of cell composition towards phenotypical stabilizing of human plaques. These stabilization features and mechanisms are directly mediated by the cells present in plaques and can be mimicked in vitro via primary plaque cells derived from human atherosclerotic tissues. In addition, the rapidly evolving of sequencing technologies identify many candidate genes and molecular mechanisms that may influence the risk of developing an atherosclerotic thrombotic event - which bring the next challenge in sharp focus: how to translate these cell-specific insights into tangible functional and translational discoveries?
Collapse
Affiliation(s)
- Michele F Buono
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Lotte Slenders
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marian Wesseling
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robin J G Hartman
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michal Mokry
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, the Netherlands; Central Diagnostics Laboratory, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
87
|
Islam S, Boström KI, Di Carlo D, Simmons CA, Tintut Y, Yao Y, Hsu JJ. The Mechanobiology of Endothelial-to-Mesenchymal Transition in Cardiovascular Disease. Front Physiol 2021; 12:734215. [PMID: 34566697 PMCID: PMC8458763 DOI: 10.3389/fphys.2021.734215] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/09/2021] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) lining the cardiovascular system are subjected to a highly dynamic microenvironment resulting from pulsatile pressure and circulating blood flow. Endothelial cells are remarkably sensitive to these forces, which are transduced to activate signaling pathways to maintain endothelial homeostasis and respond to changes in the environment. Aberrations in these biomechanical stresses, however, can trigger changes in endothelial cell phenotype and function. One process involved in this cellular plasticity is endothelial-to-mesenchymal transition (EndMT). As a result of EndMT, ECs lose cell-cell adhesion, alter their cytoskeletal organization, and gain increased migratory and invasive capabilities. EndMT has long been known to occur during cardiovascular development, but there is now a growing body of evidence also implicating it in many cardiovascular diseases (CVD), often associated with alterations in the cellular mechanical environment. In this review, we highlight the emerging role of shear stress, cyclic strain, matrix stiffness, and composition associated with EndMT in CVD. We first provide an overview of EndMT and context for how ECs sense, transduce, and respond to certain mechanical stimuli. We then describe the biomechanical features of EndMT and the role of mechanically driven EndMT in CVD. Finally, we indicate areas of open investigation to further elucidate the complexity of EndMT in the cardiovascular system. Understanding the mechanistic underpinnings of the mechanobiology of EndMT in CVD can provide insight into new opportunities for identification of novel diagnostic markers and therapeutic interventions.
Collapse
Affiliation(s)
- Shahrin Islam
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Kristina I Boström
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,UCLA Molecular Biology Institute, Los Angeles, CA, United States.,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Electrical and Computer Engineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Craig A Simmons
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON, Canada.,Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada.,Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON, Canada
| | - Yin Tintut
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Department of Physiology, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Orthopedic Surgery, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yucheng Yao
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| | - Jeffrey J Hsu
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States.,Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
88
|
Martos-Rodríguez CJ, Albarrán-Juárez J, Morales-Cano D, Caballero A, MacGrogan D, de la Pompa JL, Carramolino L, Bentzon JF. Fibrous Caps in Atherosclerosis Form by Notch-Dependent Mechanisms Common to Arterial Media Development. Arterioscler Thromb Vasc Biol 2021; 41:e427-e439. [PMID: 34261328 DOI: 10.1161/atvbaha.120.315627] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objective Atheromatous fibrous caps are produced by smooth muscle cells (SMCs) that are recruited to the subendothelial space. We tested whether the recruitment mechanisms are the same as in embryonic artery development, which relies prominently on Notch signaling to form the subendothelial medial SMC layers. Approach and Results Notch elements were expressed in regions of fibrous cap in human and mouse plaques. To assess the causal role of Notch signaling in cap formation, we studied atherosclerosis in mice where the Notch pathway was inactivated in SMCs by conditional knockout of the essential effector transcription factor RBPJ (recombination signal-binding protein for immunoglobulin kappa J region). The recruitment of cap SMCs was significantly reduced without major effects on plaque size. Lineage tracing revealed the accumulation of SMC-derived plaque cells in the cap region was unaltered but that Notch-defective cells failed to re-acquire the SMC phenotype in the cap. Conversely, to analyze whether the loss of Notch signaling is required for SMC-derived cells to accumulate in atherogenesis, we studied atherosclerosis in mice with constitutive activation of Notch signaling in SMCs achieved by conditional expression of the Notch intracellular domain. Forced Notch signaling inhibited the ability of medial SMCs to contribute to plaque cells, including both cap SMCs and osteochondrogenic cells, and significantly reduced atherosclerosis development. Conclusions Sequential loss and gain of Notch signaling is needed to build the cap SMC population. The shared mechanisms with embryonic arterial media assembly suggest that the cap forms as a neo-media that restores the connection between endothelium and subendothelial SMCs, transiently disrupted in early atherogenesis.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Arteries/metabolism
- Arteries/pathology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Lineage
- Cells, Cultured
- Disease Progression
- Fibrosis
- Humans
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics
- Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism
- Jagged-1 Protein/genetics
- Jagged-1 Protein/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Plaque, Atherosclerotic
- Rats
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
- Tunica Media/metabolism
- Tunica Media/pathology
- Mice
Collapse
Affiliation(s)
- Carlos J Martos-Rodríguez
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Julián Albarrán-Juárez
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
| | - Daniel Morales-Cano
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Ainoa Caballero
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.M., J.L.d.l.P.)
- Ciber de Enfermedades Cardiovasculares, Madrid, Spain (D.M., J.L.d.l.P.)
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain (D.M., J.L.d.l.P.)
- Ciber de Enfermedades Cardiovasculares, Madrid, Spain (D.M., J.L.d.l.P.)
| | - Laura Carramolino
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
| | - Jacob F Bentzon
- Experimental Pathology of Atherosclerosis Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (C.J.M.-R., D.M.-C., A.C., L.C., J.F.B.)
- Heart Diseases, Department of Clinical Medicine (J.A.-J., A.C., J.F.B.), Aarhus University, Denmark
- Steno Diabetes Center Aarhus, Department of Clinical Medicine (J.F.B.), Aarhus University, Denmark
| |
Collapse
|
89
|
Schlegel M, Sharma M, Brown EJ, Newman AAC, Cyr Y, Afonso MS, Corr EM, Koelwyn GJ, van Solingen C, Guzman J, Farhat R, Nikain CA, Shanley LC, Peled D, Schmidt AM, Fisher EA, Moore KJ. Silencing Myeloid Netrin-1 Induces Inflammation Resolution and Plaque Regression. Circ Res 2021; 129:530-546. [PMID: 34289717 PMCID: PMC8529357 DOI: 10.1161/circresaha.121.319313] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rationale: Therapeutic efforts to decrease atherosclerotic cardiovascular disease risk have focused largely on reducing atherogenic lipoproteins, yet lipid-lowering therapies alone are insufficient to fully regress plaque burden. We postulate that arterial repair requires resolution of a maladaptive immune response and that targeting factors that hinder inflammation resolution will facilitate plaque regression. Objective: The guidance molecule Ntn1 (netrin-1) is secreted by macrophages in atherosclerotic plaques, where it sustains inflammation by enhancing macrophage survival and blocking macrophage emigration. We tested whether silencing Ntn1 in advanced atherosclerosis could resolve arterial inflammation and regress plaques. Methods and Results: To temporally silence Ntn1 in myeloid cells, we generated genetically modified mice in which Ntn1 could be selectively deleted in monocytes and macrophages using a tamoxifen-induced CX3CR1-driven cre recombinase (Ntn1fl/flCx3cr1creERT2+) and littermate control mice (Ntn1fl/flCx3cr1WT). Mice were fed Western diet in the setting of hepatic PCSK9 (proprotein convertase subtilisin/kexin type 9) overexpression to render them atherosclerotic and then treated with tamoxifen to initiate deletion of myeloid Ntn1 (MøΔNtn1) or not in controls (MøWT). Morphometric analyses performed 4 weeks later showed that myeloid Ntn1 silencing reduced plaque burden in the aorta (−50%) and plaque complexity in the aortic root. Monocyte-macrophage tracing experiments revealed lower monocyte recruitment, macrophage retention, and proliferation in MøΔNtn1 compared with MøWT plaques, indicating a restructuring of monocyte-macrophage dynamics in the artery wall upon Ntn1 silencing. Single-cell RNA sequencing of aortic immune cells before and after Ntn1 silencing revealed upregulation of gene pathways involved in macrophage phagocytosis and migration, including the Ccr7 chemokine receptor signaling pathway required for macrophage emigration from plaques and atherosclerosis regression. Additionally, plaques from MøΔNtn1 mice showed hallmarks of inflammation resolution, including higher levels of proresolving macrophages, IL (interleukin)-10, and efferocytosis, as compared to plaques from MøWT mice. Conclusion: Our data show that targeting Ntn1 in advanced atherosclerosis ameliorates atherosclerotic inflammation and promotes plaque regression.
Collapse
Affiliation(s)
- Martin Schlegel
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
- Department of Anesthesiology and Intensive Care, Technical University of Munich, School of Medicine, Germany (M. Schlegel)
| | - Monika Sharma
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Emily J Brown
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Alexandra A C Newman
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Yannick Cyr
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Milessa Silva Afonso
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Emma M Corr
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Graeme J Koelwyn
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Coen van Solingen
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Jonathan Guzman
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Rubab Farhat
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Cyrus A Nikain
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Lianne C Shanley
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Daniel Peled
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Ann Marie Schmidt
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University (A.M.S.). K.J. Moore, M. Schlegel, M. Sharma, A.M. Schmidt, and E.A. Fisher designed the study and performed data analysis and interpretation. M. Schlegel, M. Sharma, M.S. Afonso, E.J. Brown, E.M. Corr, C. van Solingen, G.J. Koelwyn, A.A.C. Newman, Y. Cyr, R. Farhat, J. Guzman, L.C. Shanley, and D. Peled conducted experiments, acquired data, and performed analyses. E.J. Brown analyzed the RNA-sequencing data. K.J. Moore and M. Schlegel wrote the article with input from all authors
| | - Edward A Fisher
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| | - Kathryn J Moore
- NYU Cardiovascular Research Center, The Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine (M. Schlegel, M. Sharma, E.J.B., A.A.C.N., Y.C., M.S.A., E.M.C., G.J.K., C.v.S., J.G., R.F., C.A.N., L.C.S., D.P., E.A.F., K.J.M.)
| |
Collapse
|
90
|
Affiliation(s)
- Ulf Hedin
- Departments of Vascular Surgery and Molecular Medicine and Surgery, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
91
|
Childs BG, Zhang C, Shuja F, Sturmlechner I, Trewartha S, Fierro Velasco R, Baker D, Li H, van Deursen JM. Senescent cells suppress innate smooth muscle cell repair functions in atherosclerosis. NATURE AGING 2021; 1:698-714. [PMID: 34746803 PMCID: PMC8570576 DOI: 10.1038/s43587-021-00089-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Senescent cells (SNCs) degenerate the fibrous cap that normally prevents atherogenic plaque rupture, a leading cause of myocardial infarction and stroke. Here we explored the underlying mechanism using pharmacological or transgenic approaches to clear SNCs in the Ldlr -/- mouse model of atherosclerosis. SNC clearance reinforced fully deteriorated fibrous caps in highly advanced lesions, as evidenced by restored vascular smooth muscle cell (VSMC) numbers, elastin content, and overall cap thickness. We found that SNCs inhibit VSMC promigratory phenotype switching in the first interfiber space of the arterial wall directly beneath atherosclerotic plaque, thereby limiting lesion entry of medial VSMCs for fibrous cap assembly or reinforcement. SNCs do so by antagonizing IGF-1 through the secretion of insulin-like growth factor-binding protein 3 (Igfbp3). These data indicate that the intermittent use of senolytic agents or IGFBP-3 inhibition in combination with lipid lowering drugs may provide therapeutic benefit in atherosclerosis.
Collapse
Affiliation(s)
- Bennett G. Childs
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester MN, United States
| | - Fahad Shuja
- Division of Vascular and Endovascular Surgery, Mayo Clinic, Rochester MN, United States
| | - Ines Sturmlechner
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Molecular Genetics Section, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Shawn Trewartha
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Raul Fierro Velasco
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
| | - Darren Baker
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester MN, United States
| | - Jan M. van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester MN, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester MN, United States
- Correspondence:
| |
Collapse
|
92
|
Kabir I, Greif DM. SNCs meet SMCs in the atherosclerotic plaque. NATURE AGING 2021; 1:631-633. [PMID: 36540165 PMCID: PMC9762735 DOI: 10.1038/s43587-021-00096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Cellular senescence and smooth muscle cells are key features of the atherosclerotic plaque; however, how senescent cells regulate smooth muscle cells is largely unknown. Herein, a new study in Nature Aging illuminates this interplay, providing insights into plaque dynamics and stability with potentially profound implications for heart attack and stroke.
Collapse
Affiliation(s)
- Inamul Kabir
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Daniel M. Greif
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University, New Haven, CT 06511, USA
- Department of Genetics, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
93
|
Lin A, Peiris NJ, Dhaliwal H, Hakim M, Li W, Ganesh S, Ramaswamy Y, Patel S, Misra A. Mural Cells: Potential Therapeutic Targets to Bridge Cardiovascular Disease and Neurodegeneration. Cells 2021; 10:cells10030593. [PMID: 33800271 PMCID: PMC7999039 DOI: 10.3390/cells10030593] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Mural cells collectively refer to the smooth muscle cells and pericytes of the vasculature. This heterogenous population of cells play a crucial role in the regulation of blood pressure, distribution, and the structural integrity of the vascular wall. As such, dysfunction of mural cells can lead to the pathogenesis and progression of a number of diseases pertaining to the vascular system. Cardiovascular diseases, particularly atherosclerosis, are perhaps the most well-described mural cell-centric case. For instance, atherosclerotic plaques are most often described as being composed of a proliferative smooth muscle cap accompanied by a necrotic core. More recently, the role of dysfunctional mural cells in neurodegenerative diseases, such as Alzheimer’s and Parkinson’s disease, is being recognized. In this review, we begin with an exploration of the mechanisms underlying atherosclerosis and neurodegenerative diseases, such as mural cell plasticity. Next, we highlight a selection of signaling pathways (PDGF, Notch and inflammatory signaling) that are conserved across both diseases. We propose that conserved mural cell signaling mechanisms can be exploited for the identification or development of dual-pronged therapeutics that impart both cardio- and neuroprotective qualities.
Collapse
MESH Headings
- Alzheimer Disease/drug therapy
- Alzheimer Disease/genetics
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Animals
- Atherosclerosis/drug therapy
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cardiotonic Agents/pharmacology
- Disease Models, Animal
- Gene Expression Regulation
- Humans
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neuroprotective Agents/pharmacology
- Parkinson Disease/drug therapy
- Parkinson Disease/genetics
- Parkinson Disease/metabolism
- Parkinson Disease/pathology
- Pericytes/drug effects
- Pericytes/metabolism
- Pericytes/pathology
- Plaque, Atherosclerotic/drug therapy
- Plaque, Atherosclerotic/genetics
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Platelet-Derived Growth Factor/genetics
- Platelet-Derived Growth Factor/metabolism
- Receptors, Notch/genetics
- Receptors, Notch/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Alexander Lin
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Niridu Jude Peiris
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Harkirat Dhaliwal
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Maria Hakim
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Weizhen Li
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India;
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh 208016, India
| | - Yogambha Ramaswamy
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Sanjay Patel
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
- Cardiac Catheterization Laboratory, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Ashish Misra
- Heart Research Institute, Sydney, NSW 2042, Australia; (A.L.); (N.J.P.); (H.D.); (M.H.); (W.L.); (S.P.)
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence: ; Tel.: +61-18-0065-1373
| |
Collapse
|
94
|
Affiliation(s)
- Ashish Misra
- Heart Research Institute, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia.
| | - Edward A Fisher
- Department of Medicine/Division of Cardiology, New York University Grossman School of Medicine, New York, NY, USA.
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|