1
|
Armstrong SS, Chen DG, Kumar S, Heath JR, Feinstein MJ, Greenland JR, Calabrese DR, Lanier LL, Ley K, Shemesh A. CITE-Seq Analysis Reveals a Differential Natural Killer Cell SPON2 Expression in Cardiovascular Disease Patients Impacted by Human-Cytomegalovirus Serostatus and Diabetes. Int J Mol Sci 2025; 26:1369. [PMID: 39941136 PMCID: PMC11818894 DOI: 10.3390/ijms26031369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Coronary artery disease (CAD) is linked to atherosclerosis plaque formation. In pro-inflammatory conditions, human Natural Killer (NK) cell frequencies in blood or plaque decrease; however, NK cells are underexplored in CAD pathogenesis, inflammatory mechanisms, and CAD comorbidities, such as human cytomegalovirus (HCMV) infection and diabetes. Analysis of PBMC CITE-seq data from sixty-one CAD patients revealed higher blood NK cell SPON2 expression in CAD patients with higher stenosis severity. Conversely, NK cell SPON2 expression was lower in pro-inflammatory atherosclerosis plaque tissue with an enriched adaptive NK cell gene signature. In CAD patients with higher stenosis severity, peripheral blood NK cell SPON2 expression was lower in patients with high HCMV-induced adaptive NK cell frequencies and corresponded to lower PBMC TGFβ transcript expression with dependency on diabetes status. These results suggest that high NK cell SPON2 expression is linked to atherosclerosis pro-homeostatic status and may have diagnostic and prognostic implications in cardiovascular disease.
Collapse
Affiliation(s)
| | - Daniel G. Chen
- Institute of Systems Biology, University of Washington, Seattle, WA 98109, USA; (D.G.C.)
| | - Sunil Kumar
- Immunology Center of Georgia, Medical College of Georgia, Augusta, GA 30912, USA
| | - James R. Heath
- Institute of Systems Biology, University of Washington, Seattle, WA 98109, USA; (D.G.C.)
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA;
| | - Matthew J. Feinstein
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - John R. Greenland
- Department of Medicine, University of California, San Francisco, CA 94143, USA
- Medical Service, VA Health Care System, San Francisco, CA 94121, USA
| | - Daniel R. Calabrese
- Department of Medicine, University of California, San Francisco, CA 94143, USA
- Medical Service, VA Health Care System, San Francisco, CA 94121, USA
| | - Lewis L. Lanier
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA;
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | - Klaus Ley
- Immunology Center of Georgia, Medical College of Georgia, Augusta, GA 30912, USA
| | - Avishai Shemesh
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA;
- Department of Medicine, University of California, San Francisco, CA 94143, USA
- Medical Service, VA Health Care System, San Francisco, CA 94121, USA
| |
Collapse
|
2
|
Lu Y, Wang Y, Ruan T, Wang Y, Ju L, Zhou M, Liu L, Yao D, Yao M. Immunometabolism of Tregs: mechanisms, adaptability, and therapeutic implications in diseases. Front Immunol 2025; 16:1536020. [PMID: 39917294 PMCID: PMC11798928 DOI: 10.3389/fimmu.2025.1536020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Immunometabolism is an emerging field that explores the intricate interplay between immune cells and metabolism. Regulatory T cells (Tregs), which maintain immune homeostasis in immunometabolism, play crucial regulatory roles. The activation, differentiation, and function of Tregs are influenced by various metabolic pathways, such as the Mammalian targets of rapamycin (mTOR) pathway and glycolysis. Correspondingly, activated Tregs can reciprocally impact these metabolic pathways. Tregs also possess robust adaptive capabilities, thus enabling them to adapt to various microenvironments, including the tumor microenvironment (TME). The complex mechanisms of Tregs in metabolic diseases are intriguing, particularly in conditions like MASLD, where Tregs are significantly upregulated and contribute to fibrosis, while in diabetes, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA), they show downregulation and reduced anti-inflammatory capacity. These phenomena suggest that the differentiation and function of Tregs are influenced by the metabolic environment, and imbalances in either can lead to the development of metabolic diseases. Thus, moderate differentiation and inhibitory capacity of Tregs are critical for maintaining immune system balance. Given the unique immunoregulatory abilities of Tregs, the development of targeted therapeutic drugs may position them as novel targets in immunotherapy. This could contribute to restoring immune system balance, resolving metabolic dysregulation, and fostering innovation and progress in immunotherapy.
Collapse
|
3
|
Pessentheiner AR, Spann NJ, Autran CA, Oh TG, Grunddal KV, Coker JK, Painter CD, Ramms B, Chiang AW, Wang CY, Hsiao J, Wang Y, Quach A, Booshehri LM, Hammond A, Tognaccini C, Latasiewicz J, Willemsen L, Zengler K, de Winther MP, Hoffman HM, Philpott M, Cribbs AP, Oppermann U, Lewis NE, Witztum JL, Yu R, Atkins AR, Downes M, Evans RM, Glass CK, Bode L, Gordts PL. The human milk oligosaccharide 3'sialyllactose reduces low-grade inflammation and atherosclerosis development in mice. JCI Insight 2024; 9:e181329. [PMID: 39325548 PMCID: PMC11601559 DOI: 10.1172/jci.insight.181329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024] Open
Abstract
Macrophages contribute to the induction and resolution of inflammation and play a central role in chronic low-grade inflammation in cardiovascular diseases caused by atherosclerosis. Human milk oligosaccharides (HMOs) are complex unconjugated glycans unique to human milk that benefit infant health and act as innate immune modulators. Here, we identify the HMO 3'sialyllactose (3'SL) as a natural inhibitor of TLR4-induced low-grade inflammation in macrophages and endothelium. Transcriptome analysis in macrophages revealed that 3'SL attenuates mRNA levels of a selected set of inflammatory genes and promotes the activity of liver X receptor (LXR) and sterol regulatory element binding protein-1 (SREBP1). These acute antiinflammatory effects of 3'SL were associated with reduced histone H3K27 acetylation at a subset of LPS-inducible enhancers distinguished by preferential enrichment for CCCTC-binding factor (CTCF), IFN regulatory factor 2 (IRF2), B cell lymphoma 6 (BCL6), and other transcription factor recognition motifs. In a murine atherosclerosis model, both s.c. and oral administration of 3'SL significantly reduced atherosclerosis development and the associated inflammation. This study provides evidence that 3'SL attenuates inflammation by a transcriptional mechanism to reduce atherosclerosis development in the context of cardiovascular disease.
Collapse
Affiliation(s)
- Ariane R. Pessentheiner
- Department of Medicine, UCSD, La Jolla, California, USA
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | | | | | - Tae Gyu Oh
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | | | - Joanna K.C. Coker
- Department of Medicine, UCSD, La Jolla, California, USA
- Department of Bioengineering at UCSD, La Jolla, California, USA
| | | | - Bastian Ramms
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Austin W.T. Chiang
- Department of Pediatrics at UCSD, La Jolla, California, USA
- Department of Bioengineering at UCSD, La Jolla, California, USA
- Novo Nordisk Foundation Center for Biosustainability, La Jolla, California, USA
| | - Chen-Yi Wang
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
- Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, United Kingdom
| | - Jason Hsiao
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Yiwen Wang
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Anthony Quach
- Department of Medicine, UCSD, La Jolla, California, USA
| | | | | | | | | | - Lisa Willemsen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Karsten Zengler
- Department of Bioengineering at UCSD, La Jolla, California, USA
- Center for Microbiome Innovation, UCSD, La Jolla, California, USA
| | - Menno P.J. de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Hal M. Hoffman
- Department of Medicine, UCSD, La Jolla, California, USA
- Department of Pediatrics at UCSD, La Jolla, California, USA
- Rady Children’s Hospital of San Diego, San Diego, California, USA
| | - Martin Philpott
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
| | - Adam P. Cribbs
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
- Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, United Kingdom
| | - Udo Oppermann
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, NIH Research Oxford Biomedical Research Unit (BRU), and
- Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, United Kingdom
| | - Nathan E. Lewis
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
- Department of Bioengineering at UCSD, La Jolla, California, USA
- Novo Nordisk Foundation Center for Biosustainability, La Jolla, California, USA
| | | | - Ruth Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Annette R. Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Ron M. Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Christopher K. Glass
- Department of Medicine, UCSD, La Jolla, California, USA
- Department of Cellular and Molecular Medicine and
| | - Lars Bode
- Department of Pediatrics at UCSD, La Jolla, California, USA
- Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE) and
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| | - Philip L.S.M. Gordts
- Department of Medicine, UCSD, La Jolla, California, USA
- Glycobiology Research and Training Center, UCSD, La Jolla, California, USA
| |
Collapse
|
4
|
Yang Q, Liu J, Zhang T, Zhu T, Yao S, Wang R, Wang W, Dilimulati H, Ge J, An S. Exploring shared biomarkers and shared pathways in insomnia and atherosclerosis using integrated bioinformatics analysis. Front Mol Neurosci 2024; 17:1477903. [PMID: 39439987 PMCID: PMC11493776 DOI: 10.3389/fnmol.2024.1477903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background Insomnia (ISM) is one of the non-traditional drivers of atherosclerosis (AS) and an important risk factor for AS-related cardiovascular disease. Our study aimed to explore the shared pathways and diagnostic biomarkers of ISM-related AS using integrated bioinformatics analysis. Methods We download the datasets from the Gene Expression Omnibus database and the GeneCards database. Weighted gene co-expression network analysis and gene differential expression analysis were applied to screen the AS-related gene set. The shared genes of ISM and AS were obtained by intersecting with ISM-related genes. Subsequently, candidate diagnostic biomarkers were identified by constructing protein-protein interaction networks and machine learning algorithms, and a nomogram was constructed. Moreover, to explore potential mechanisms, a comprehensive analysis of shared genes was carried out, including enrichment analysis, protein interactions, immune cell infiltration, and single-cell sequencing analysis. Results We successfully screened 61 genes shared by ISM and AS, of which 3 genes (IL10RA, CCR1, and SPI1) were identified as diagnostic biomarkers. A nomogram with excellent predictive value was constructed (the area under curve of the model constructed by the biomarkers was 0.931, and the validation set was 0.745). In addition, the shared genes were mainly enriched in immune and inflammatory response regulation pathways. The biomarkers were associated with a variety of immune cells, especially myeloid immune cells. Conclusion We constructed a diagnostic nomogram based on IL10RA, CCR1, and SPI1 and explored the inflammatory-immune mechanisms, which indicated new insights for early diagnosis and treatment of ISM-related AS.
Collapse
Affiliation(s)
- Qichong Yang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Juncheng Liu
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
- Henan Province People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingting Zhang
- Center for Clinical Single-Cell Biomedicine, Henan Province People’s Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingting Zhu
- Department of Cardiopulmonary Functions Test, Henan Province People’s Hospital, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Siyu Yao
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Rongzi Wang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Wenjuan Wang
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Haliminai Dilimulati
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Songtao An
- Central China Fuwai Hospital of Zhengzhou University, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Regenerative Medicine, National Health Commission, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Kerns S, Owen KA, Daamen A, Kain J, Grammer AC, Lipsky PE. Genetic association with autoimmune diseases identifies molecular mechanisms of coronary artery disease. iScience 2024; 27:110715. [PMID: 39262791 PMCID: PMC11387803 DOI: 10.1016/j.isci.2024.110715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/28/2024] [Accepted: 08/08/2024] [Indexed: 09/13/2024] Open
Abstract
Autoimmune patients have a significantly increased risk of developing coronary artery disease (CAD) compared to the general population. However, autoimmune patients often lack traditional risk factors for CAD and there is increasing recognition of inflammation in CAD development. In this study, we leveraged genome-wide association study (GWAS) data to understand whether there is a genetic relationship between CAD and autoimmunity. Statistical genetic comparison methods were used to identify correlated and causal SNPs between various autoimmune diseases and CAD. Pleiotropic SNPs were identified by cross-phenotype association analysis (CPASSOC) and overlap between GWAS. Causal SNPs were identified using Mendelian Randomization (MR) and Colocalization (COLOC). Using SNP-to-gene mapping, we additionally identified pleiotropic and causal genes and pathways associated between autoimmunity and CAD, which were contextualized by documentation of enrichment in individual cell types identified from coronary atherosclerotic plaques by single-cell RNA sequencing. These results provide insight into potential inflammatory therapeutic targets for CAD.
Collapse
Affiliation(s)
- Sophia Kerns
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Katherine A Owen
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Andrea Daamen
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Jessica Kain
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
- Stanford University Department of Genetics, Stanford, CA 94305, USA
| | - Amrie C Grammer
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| | - Peter E Lipsky
- AMPEL Biosolutions, LLC, Charlottesville, VA 22903, USA
- The RILITE Research Institute, Charlottesville, VA 22903, USA
| |
Collapse
|
6
|
Liu Y, Wang Z, Fang L, Xu Y, Zhao B, Kang X, Zhao Y, Han J, Zhang Y, Dong E, Wang N. Deficiency of 5-HT 2B receptors alleviates atherosclerosis by regulating macrophage phenotype through inhibiting interferon signalling. Br J Pharmacol 2024. [PMID: 39232850 DOI: 10.1111/bph.17315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/09/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND AND PURPOSE Elevated levels of 5-HT have been correlated with coronary artery disease and cardiac events, suggesting 5-HT is a potential novel factor in the development of atherosclerotic cardiovascular disease. However, the underlying pathological mechanisms of the 5-HT system in atherosclerosis remain unclear. The 5-HT2B receptor (5-HT2BR), which establishes a positive feedback loop with 5-HT, has been identified as a contributor to pathophysiological processes in various vascular disorders. In this study, we investigated the immunological impact of 5-HT2BR in atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice. EXPERIMENTAL APPROACH Plasma levels of 5-HT were measured in mice using an ELISA kit. Atherosclerotic plaque formation, macrophage infiltration and inflammatory signalling were assessed in ApoE-/- mice by employing both pharmacological inhibition and genetic deficiency of 5-HT2BR. Inflammasome activation was elucidated using peritoneal macrophages isolated from 5-HT2BR-deficient mice. KEY RESULTS An upregulation of 5-HT2BR expression was observed in the aortas of ApoE-/- mice, exhibiting a strong correlation with the presence of macrophages in plaques. Atherosclerosis was attenuated in mice through pharmacological inhibition and genetic deficiency of 5-HT2BR. Additionally, a significant reduction in atherosclerotic plaque size was achieved through bone marrow reconstitution with 5-HT2BR-deficient cells. 5-HT2BR-deficient macrophages showed attenuated interferon (IFN) signalling, NLRP3 inflammasome activation, and interleukin-1β release. Moreover, macrophages primed with 5-HT2BR deficiency displayed an anti-inflammatory phenotype. CONCLUSION AND IMPLICATIONS These findings support the hypothesis that 5-HT2BR in macrophages plays a causal role in the development of atherosclerosis, revealing a novel perspective for potential therapeutic strategies in atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Yahan Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhipeng Wang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Fang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yaohua Xu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Beilei Zhao
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xuya Kang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanqing Zhao
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Jintao Han
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Institute of Cardiovascular Diseases, The first affiliated Hospital of Dalian Medical University, Dalian, China
| | - Erdan Dong
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital); School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Nanping Wang
- Wuhu Hospital, East China Normal University (ECNU), Wuhu, China
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|
7
|
Lorentsson HJN, Clausen CR, Faurholt-Jepsen D, Hansen KB, Jensen SG, Krogh-Madsen R, Hagelqvist PG, Johansson PI, Vilsbøll T, Knop FK, Ravn P. The effect of Mycobacterium tuberculosis treatment on thrombelastography-assessed haemostasis: a prospective cohort study. Thromb J 2024; 22:54. [PMID: 38918780 PMCID: PMC11201340 DOI: 10.1186/s12959-024-00625-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Tuberculosis disease (TB) and tuberculosis infection (TBI) have been associated with increased risk of cardiovascular disease which may be connected to infection-related haemostatic changes. It is unknown if treatment of Mycobacterium tuberculosis influences haemostasis. Here, we assessed if TB or TBI treatment affects thrombelastography (TEG)-assessed haemostasis. METHODS Individuals with TB or TBI were included from a TB outpatient clinic in Copenhagen, Denmark. Patients treated with antithrombotic medication or systemic immunosuppressants were excluded. TEG analysis was performed before and after TB/TBI treatment using the TEG®6s analyser to provide data on the reaction time of clot initiation (R) (min), the speed of clot formation (K) (min) and clot build-up (Angle) (°), maximum clot strength (MA) (mm), and clot breakdown/fibrinolysis (LY30) (%). Differences in TEG were assessed using paired t tests. RESULTS We included eleven individuals with TB with median [interquartile range] [IQR] age 52 (Liu et al. in Medicine (United States) 95, 2016) years and mean (standard deviation) (SD) body mass index (BMI) 24.7 (6.3) kg/m2 as well as 15 individuals with TBI with median [IQR] age 49 (Wells et al. in Am J Respir Crit Care Med 204:583, 2021) years and BMI 26.0 (3.2) kg/m2. Treatment reduced MA for both TB (64.0 (6.3) vs. 57.9 (5.2) mm, p = 0.016) and TBI (61.3 (4.1) vs. 58.6 (5.0) mm, p = 0.023) whereas R, K, Angle and LY30 were unaffected. CONCLUSION TEG analysis showed that treatments of TB and TBI were associated with reduced MA which may indicate the existence of cardiovascular benefits from therapy. TRIAL REGISTRATION Registered at ClinicalTrials.gov 05 April 2021 with registration number NCT04830462.
Collapse
Affiliation(s)
- Hans Johan Niklas Lorentsson
- Section of Infectious Diseases, Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark.
- Section of Infectious Diseases, Department of Medicine, Herlev and Gentofte Hospital, Hellerup Hospitalsvej 1, +45 38 67 38 67, Hellerup, 2900, Denmark.
| | - Christina R Clausen
- Section of Infectious Diseases, Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Daniel Faurholt-Jepsen
- Department of Infectious Diseases, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Sidse Graff Jensen
- Section of Respiratory Diseases, Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Rikke Krogh-Madsen
- Department of Infectious Diseases, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
- Center for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Per G Hagelqvist
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Pär I Johansson
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- CAG Center for Endotheliomics, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pernille Ravn
- Section of Infectious Diseases, Department of Medicine, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Song L, Zhang B, Li R, Duan Y, Chi Y, Xu Y, Hua X, Xu Q. Significance of neutrophil extracellular traps-related gene in the diagnosis and classification of atherosclerosis. Apoptosis 2024; 29:605-619. [PMID: 38367202 DOI: 10.1007/s10495-023-01923-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2023] [Indexed: 02/19/2024]
Abstract
Atherosclerosis (AS) is a pathological process associated with various cardiovascular diseases. Upon different stimuli, neutrophils release reticular complexes known as neutrophil extracellular traps (NETs). Numerous researches have indicated a strong correlation between NETs and AS. However, its role in cardiovascular disease requires further investigation. By utilizing a machine learning algorithm, we examined the genes associated with NETs that were expressed differently in individuals with AS compared to normal controls. As a result, we identified four distinct genes. A nomogram model was built to forecast the incidence of AS. Additionally, we conducted analysis on immune infiltration, functional enrichment and consensus clustering in AS samples. The findings indicated that individuals with AS could be categorized into two groups, exhibiting notable variations in immune infiltration traits among the groups. Furthermore, to measure the NETs model, the principal component analysis algorithm was developed and cluster B outperformed cluster A in terms of NETs. Additionally, there were variations in the expression of multiple chemokines between the two subtypes. By studying AS NETs, we acquired fresh knowledge about the molecular patterns and immune mechanisms implicated, which could open up new possibilities for AS immunotherapy.
Collapse
Affiliation(s)
- Liantai Song
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Boyu Zhang
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Reng Li
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Yibing Duan
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Yifan Chi
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Yangyi Xu
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Xucong Hua
- Basic Medical College of Chengde Medical University, Chengde, 067000, China
| | - Qian Xu
- Department of Biochemistry, Chengde Medical University, Chengde, 067000, Hebei, People's Republic of China.
| |
Collapse
|
9
|
Jansen I, Cahalane R, Hengst R, Akyildiz A, Farrell E, Gijsen F, Aikawa E, van der Heiden K, Wissing T. The interplay of collagen, macrophages, and microcalcification in atherosclerotic plaque cap rupture mechanics. Basic Res Cardiol 2024; 119:193-213. [PMID: 38329498 PMCID: PMC11008085 DOI: 10.1007/s00395-024-01033-5] [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: 08/21/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
The rupture of an atherosclerotic plaque cap overlying a lipid pool and/or necrotic core can lead to thrombotic cardiovascular events. In essence, the rupture of the plaque cap is a mechanical event, which occurs when the local stress exceeds the local tissue strength. However, due to inter- and intra-cap heterogeneity, the resulting ultimate cap strength varies, causing proper assessment of the plaque at risk of rupture to be lacking. Important players involved in tissue strength include the load-bearing collagenous matrix, macrophages, as major promoters of extracellular matrix degradation, and microcalcifications, deposits that can exacerbate local stress, increasing tissue propensity for rupture. This review summarizes the role of these components individually in tissue mechanics, along with the interplay between them. We argue that to be able to improve risk assessment, a better understanding of the effect of these individual components, as well as their reciprocal relationships on cap mechanics, is required. Finally, we discuss potential future steps, including a holistic multidisciplinary approach, multifactorial 3D in vitro model systems, and advancements in imaging techniques. The obtained knowledge will ultimately serve as input to help diagnose, prevent, and treat atherosclerotic cap rupture.
Collapse
Affiliation(s)
- Imke Jansen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rachel Cahalane
- Mechanobiology and Medical Device Research Group (MMDRG), Biomedical Engineering, College of Science and Engineering, University of Galway, Galway, Ireland
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ranmadusha Hengst
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ali Akyildiz
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Frank Gijsen
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Biomechanical Engineering, Technical University Delft, Delft, The Netherlands
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kim van der Heiden
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Wissing
- Department of Biomedical Engineering, Thorax Center Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
10
|
Song YJ, Ma Y, Meng T, Zhuang T, Ruan CC, Li Y, Zhang GN. The Characteristics of Macrophage Heterogeneity in Atherosclerotic Aortas. J Cardiovasc Transl Res 2024; 17:153-166. [PMID: 37713049 DOI: 10.1007/s12265-023-10434-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023]
Abstract
Macrophage is the main effector cell during atherosclerosis. We applied single-cell RNA sequencing (scRNA) data to investigate the role of macrophage subsets in atherosclerosis. Monocyte and macrophage clusters were divided into 6 subclusters. Each subcluster's markers were calculated and validated by immunofluorescence. Elevated macrophage subclusters in the WD group were subject to enrichment pathway analysis and exhibited different phenotypes. Pseudotime analysis shows the subclusters originate from monocytes. We cultured bone marrow-derived macrophages with CSF-1 and ox-LDL to simulate an atherosclerotic-like environment and detected the transformation of subclusters. Macrophage-Vegfa and Macrophage-C1qb increased in the WD group. Macrophage-Vegfa acquires the characteristics of phagocytosis and immune response, while Macrophage-C1qb is not involved in lipid metabolism. The two subclusters are both enriched in cell movement and migration pathways. Experimental verification proved Monocyte-Ly6C evolved into Macrophage-Vegfa and Macrophage-C1qb during atherosclerosis progression.
Collapse
Affiliation(s)
- Yu-Jie Song
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Ma
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Guan-Nan Zhang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
Wieland EB, Kempen LJ, Donners MM, Biessen EA, Goossens P. Macrophage heterogeneity in atherosclerosis: A matter of context. Eur J Immunol 2024; 54:e2350464. [PMID: 37943053 DOI: 10.1002/eji.202350464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/19/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023]
Abstract
During atherogenesis, plaque macrophages take up and process deposited lipids, trigger inflammation, and form necrotic cores. The traditional inflammatory/anti-inflammatory paradigm has proven insufficient in explaining their complex disease-driving mechanisms. Instead, we now appreciate that macrophages exhibit remarkable heterogeneity and functional specialization in various pathological contexts, including atherosclerosis. Technical advances for studying individual cells, especially single-cell RNA sequencing, indeed allowed to identify novel macrophage subsets in both murine and human atherosclerosis, highlighting the existence of diverse macrophage activation states throughout pathogenesis. In addition, recent studies highlighted the role of the local microenvironment in shaping the macrophages' phenotype and function. However, this remains largely undescribed in the context of atherosclerosis. In this review we explore the origins of macrophages and their functional specialization, shedding light on the diverse sources of macrophage accumulation in the atherosclerotic plaque. Next, we discuss the phenotypic diversity observed in both murine and human atherosclerosis, elucidating their distinct functions and spatial distribution within plaques. Finally, we highlight the importance of the local microenvironment in both phenotypic and functional specialization of macrophages in atherosclerosis and elaborate on the need for spatial multiomics approaches to provide a better understanding of the different macrophage subsets' roles in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Elias B Wieland
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Laura Jap Kempen
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- Laboratory of Immunology and Vaccinology, Faculty of Veterinary Medicine, FARAH, ULiège, Liège, Belgium
- Laboratory of Immunophysiology, GIGA Institute, Liege University, Liège, Belgium
| | - Marjo Mpc Donners
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Erik Al Biessen
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | - Pieter Goossens
- Cardiovascular Research Institute Maastricht, Experimental Vascular Pathology, Department of Pathology, Maastricht University Medical Centre+, Maastricht, the Netherlands
| |
Collapse
|
12
|
Chan A, Torelli S, Cheng E, Batchelder R, Waliany S, Neal J, Witteles R, Nguyen P, Cheng P, Zhu H. Immunotherapy-Associated Atherosclerosis: A Comprehensive Review of Recent Findings and Implications for Future Research. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:715-735. [PMID: 38213548 PMCID: PMC10776491 DOI: 10.1007/s11936-023-01024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 01/13/2024]
Abstract
Purpose of the Review Even as immune checkpoint inhibitors (ICIs) have transformed the lifespan of many patients, they may also trigger acceleration of long-term cardiovascular disease. Our review aims to examine the current landscape of research on ICI-mediated atherosclerosis and address key questions regarding its pathogenesis and impact on patient management. Recent Findings Preclinical mouse models suggest that T cell dysregulation and proatherogenic cytokine production are key contributors to plaque development after checkpoint inhibition. Clinical data also highlight the significant burden of atherosclerotic cardiovascular disease (ASCVD) in patients on immunotherapy, although the value of proactively preventing and treating ASCVD in this population remains an open area of inquiry. Current treatment options include dietary/lifestyle modification and traditional medications to manage hypertension, hyperlipidemia, and diabetes risk factors; no current targeted therapies exist. Summary Early identification of high-risk patients is crucial for effective preventive strategies and timely intervention. Future research should focus on refining screening tools, elucidating targetable mechanisms driving ICI atherosclerosis, and evaluating long-term cardiovascular outcomes in cancer survivors who received immunotherapy. Moreover, close collaboration between oncologists and cardiologists is essential to optimize patient outcomes.
Collapse
Affiliation(s)
- Antonia Chan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Evaline Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ryan Batchelder
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Paul Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| |
Collapse
|
13
|
Rioux B, Chong M, Walker R, McGlasson S, Rannikmäe K, McCartney D, McCabe J, Brown R, Crow YJ, Hunt D, Whiteley W. Phenotypes associated with genetic determinants of type I interferon regulation in the UK Biobank: a protocol. Wellcome Open Res 2023; 8:550. [PMID: 38855722 PMCID: PMC11162527 DOI: 10.12688/wellcomeopenres.20385.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2023] [Indexed: 06/11/2024] Open
Abstract
Background Type I interferons are cytokines involved in innate immunity against viruses. Genetic disorders of type I interferon regulation are associated with a range of autoimmune and cerebrovascular phenotypes. Carriers of pathogenic variants involved in genetic disorders of type I interferons are generally considered asymptomatic. Preliminary data suggests, however, that genetically determined dysregulation of type I interferon responses is associated with autoimmunity, and may also be relevant to sporadic cerebrovascular disease and dementia. We aim to determine whether functional variants in genes involved in type I interferon regulation and signalling are associated with the risk of autoimmunity, stroke, and dementia in a population cohort. Methods We will perform a hypothesis-driven candidate pathway association study of type I interferon-related genes using rare variants in the UK Biobank (UKB). We will manually curate type I interferon regulation and signalling genes from a literature review and Gene Ontology, followed by clinical and functional filtering. Variants of interest will be included based on pre-defined clinical relevance and functional annotations (using LOFTEE, M-CAP and a minor allele frequency <0.1%). The association of variants with 15 clinical and three neuroradiological phenotypes will be assessed with a rare variant genetic risk score and gene-level tests, using a Bonferroni-corrected p-value threshold from the number of genetic units and phenotypes tested. We will explore the association of significant genetic units with 196 additional health-related outcomes to help interpret their relevance and explore the clinical spectrum of genetic perturbations of type I interferon. Ethics and dissemination The UKB has received ethical approval from the North West Multicentre Research Ethics Committee, and all participants provided written informed consent at recruitment. This research will be conducted using the UKB Resource under application number 93160. We expect to disseminate our results in a peer-reviewed journal and at an international cardiovascular conference.
Collapse
Affiliation(s)
- Bastien Rioux
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Michael Chong
- Population Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, McMaster University, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Rosie Walker
- Department of Psychology, University of Exeter, Exeter, England, UK
| | - Sarah McGlasson
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Kristiina Rannikmäe
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, Scotland, UK
| | - Daniel McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
| | - John McCabe
- School of Medicine, University College Dublin, Dublin, Leinster, Ireland
- Department of Medicine for the Elderly, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Robin Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, UK
| | - Yanick J. Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, UK
- Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Université de Paris, Paris, France
| | - David Hunt
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - William Whiteley
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
- MRC Population Health Unit, Nuffield Department of Population Health, University of Oxford, Oxford, England, UK
| |
Collapse
|
14
|
Lorey MB, Youssef A, Äikäs L, Borrelli M, Hermansson M, Assini JM, Kemppainen A, Ruhanen H, Ruuth M, Matikainen S, Kovanen PT, Käkelä R, Boffa MB, Koschinsky ML, Öörni K. Lipoprotein(a) induces caspase-1 activation and IL-1 signaling in human macrophages. Front Cardiovasc Med 2023; 10:1130162. [PMID: 37293282 PMCID: PMC10244518 DOI: 10.3389/fcvm.2023.1130162] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 05/02/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction Lipoprotein(a) (Lp(a)) is an LDL-like particle with an additional apolipoprotein (apo)(a) covalently attached. Elevated levels of circulating Lp(a) are a risk factor for atherosclerosis. A proinflammatory role for Lp(a) has been proposed, but its molecular details are incompletely defined. Methods and results To explore the effect of Lp(a) on human macrophages we performed RNA sequencing on THP-1 macrophages treated with Lp(a) or recombinant apo(a), which showed that especially Lp(a) induces potent inflammatory responses. Thus, we stimulated THP-1 macrophages with serum containing various Lp(a) levels to investigate their correlations with cytokines highlighted by the RNAseq, showing significant correlations with caspase-1 activity and secretion of IL-1β and IL-18. We further isolated both Lp(a) and LDL particles from three donors and then compared their atheroinflammatory potentials together with recombinant apo(a) in primary and THP-1 derived macrophages. Compared with LDL, Lp(a) induced a robust and dose-dependent caspase-1 activation and release of IL-1β and IL-18 in both macrophage types. Recombinant apo(a) strongly induced caspase-1 activation and IL-1β release in THP-1 macrophages but yielded weak responses in primary macrophages. Structural analysis of these particles revealed that the Lp(a) proteome was enriched in proteins associated with complement activation and coagulation, and its lipidome was relatively deficient in polyunsaturated fatty acids and had a high n-6/n-3 ratio promoting inflammation. Discussion Our data show that Lp(a) particles induce the expression of inflammatory genes, and Lp(a) and to a lesser extent apo(a) induce caspase-1 activation and IL-1 signaling. Major differences in the molecular profiles between Lp(a) and LDL contribute to Lp(a) being more atheroinflammatory.
Collapse
Affiliation(s)
- Martina B. Lorey
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Amer Youssef
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Lauri Äikäs
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Matthew Borrelli
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Martin Hermansson
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Julia M. Assini
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Aapeli Kemppainen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Hanna Ruhanen
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Helsinki, Finland
| | - Maija Ruuth
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Sampsa Matikainen
- Helsinki Rheumatic Disease and Inflammation Research Group, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Reijo Käkelä
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Helsinki University Lipidomics Unit (HiLIPID), Helsinki Institute of Life Science (HiLIFE) and Biocenter Finland, Helsinki, Finland
| | - Michael B. Boffa
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Marlys L. Koschinsky
- Robarts Research Institute, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON, Canada
| | - Katariina Öörni
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
15
|
Ng CT, Fong LY, Abdullah MNH. Interferon-gamma (IFN-γ): Reviewing its mechanisms and signaling pathways on the regulation of endothelial barrier function. Cytokine 2023; 166:156208. [PMID: 37088004 DOI: 10.1016/j.cyto.2023.156208] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Abstract
Interferon-gamma (IFN-γ) is a pleiotropic cytokine that plays a critical role in mediating an array of immune responses including promotes antiviral activity, facilitates macrophage activation, controls Th1/Th2 balance, and regulates cellular apoptosis and proliferation. A few articles have previously reviewed the effects of IFN-γ in the regulation of barrier permeability, but none of these articles focuses on barrier function of endothelial cells. This review aims to discuss the regulatory mechanisms of IFN-γ on endothelial barrier function and its underlying signaling pathways. Articles were retrieved from electronic databases such as PubMed and Google Scholar using keywords "Interferon-gamma", "endothelial cells", "barrier function", and "signaling pathway". The articles published between 2000 and 2022 that are related to the aforementioned topics were selected. A few journals published beyond this period were also included due to limited information available. The results showed that IFN-γ modulates endothelial barrier function, mainly involves small GTPases, STAT1-dependent pathway, p38 MAPK and nitric oxide. In conclusion, more in depth cellular and molecular studies are needed to elucidate the pathways of IFN-γ in the regulation of endothelial barrier function.
Collapse
Affiliation(s)
- Chin Theng Ng
- Unit of Physiology, Faculty of Medicine, AIMST University, Bedong, 08100 Kedah, Malaysia.
| | - Lai Yen Fong
- Department of Pre-clinical Sciences, M. Kandiah Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Kajang, 43000 Selangor, Malaysia
| | - Muhammad Nazrul Hakim Abdullah
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, 43400 Selangor, Malaysia
| |
Collapse
|
16
|
Yang S, Zhao M, Jia S. Macrophage: Key player in the pathogenesis of autoimmune diseases. Front Immunol 2023; 14:1080310. [PMID: 36865559 PMCID: PMC9974150 DOI: 10.3389/fimmu.2023.1080310] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 02/16/2023] Open
Abstract
The macrophage is an essential part of the innate immune system and also serves as the bridge between innate immunity and adaptive immune response. As the initiator and executor of the adaptive immune response, macrophage plays an important role in various physiological processes such as immune tolerance, fibrosis, inflammatory response, angiogenesis and phagocytosis of apoptotic cells. Consequently, macrophage dysfunction is a vital cause of the occurrence and development of autoimmune diseases. In this review, we mainly discuss the functions of macrophages in autoimmune diseases, especially in systemic lupus erythematosus (SLE), rheumatic arthritis (RA), systemic sclerosis (SSc) and type 1 diabetes (T1D), providing references for the treatment and prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Shuang Yang
- Dapartment of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Zhao
- Dapartment of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.,Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
17
|
Liu C, Zhou Y, Zhou Y, Tang X, Tang L, Wang J. Identification of crucial genes for predicting the risk of atherosclerosis with system lupus erythematosus based on comprehensive bioinformatics analysis and machine learning. Comput Biol Med 2023; 152:106388. [PMID: 36470144 DOI: 10.1016/j.compbiomed.2022.106388] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) has become a major public health problem over the years, and atherosclerosis (AS) is one of the main complications of SLE associated with serious cardiovascular consequences in this patient population. The present study aimed to identify potential biomarkers for SLE patients with AS. METHODS Five microarray datasets (GSE50772, GSE81622, GSE100927, GSE28829, GSE37356) were downloaded from the NCBI Gene Expression Omnibus database. The Limma package was used to identify differentially expressed genes (DEGs) in AS. Weighted gene coexpression network analysis (WGCNA) was used to identify significant module genes associated with SLE. Functional enrichment analysis, protein-protein interaction (PPI) network construction, and machine learning algorithms (least absolute shrinkage and selection operator (Lasso, Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and random forest) were applied to identify hub genes. Subsequently, we generated a nomogram and receiver operating characteristic curve (ROC) for predicting the risk of AS in SLE patients. Finally, immune cell infiltrations were analyzed, and Consensus Cluster Analysis was conducted based on Single Sample Gene Set Enrichment Analysis (ssGSEA) scores. RESULTS Five hub genes (SPI1, MMP9, C1QA, CX3CR1, and MNDA) were identified and used to establish a nomogram that yielded a high predictive performance (area under the curve 0.900-0.981). Dysregulated immune cell infiltrations were found in AS, with positive correlations with the five hub genes. Consensus clustering showed that the optimal number of subtypes was 3. Compared to subtypes A and B, subtype C presented higher expression of the five hub genes, immune cell infiltration levels and immune checkpoint expression. CONCLUSION Our study systematically identified five candidate hub genes (SPI1, MMP9, C1QA, CX3CR1, MNDA) and established a nomogram that could predict the risk of AS with SLE using various bioinformatic analyses and machine learning algorithms. Our findings provide the foothold for future studies on potential crucial genes for AS in SLE patients. Additionally, the dysregulated immune cell proportions and immune checkpoint expressions in AS with SLE were identified.
Collapse
Affiliation(s)
- Chunjiang Liu
- Department of General Surgery, Division of Vascular Surgery, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, 312000, China
| | - Yufei Zhou
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yue Zhou
- Department of General Surgery, Division of Vascular Surgery, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, 312000, China
| | - Xiaoqi Tang
- Department of General Surgery, Division of Vascular Surgery, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, 312000, China
| | - Liming Tang
- Department of General Surgery, Division of Vascular Surgery, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, 312000, China.
| | - Jiajia Wang
- Department of Rheumatology, Shaoxing People's Hospital (Shaoxing Hospital of Zhejiang University), Shaoxing, 312000, China.
| |
Collapse
|
18
|
Tsioufis P, Theofilis P, Tsioufis K, Tousoulis D. The Impact of Cytokines in Coronary Atherosclerotic Plaque: Current Therapeutic Approaches. Int J Mol Sci 2022; 23:15937. [PMID: 36555579 PMCID: PMC9788180 DOI: 10.3390/ijms232415937] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronary atherosclerosis is a chronic pathological process that involves inflammation together with endothelial dysfunction and lipoprotein dysregulation. Experimental studies during the past decades have established the role of inflammatory cytokines in coronary artery disease, namely interleukins (ILs), tumor necrosis factor (TNF)-α, interferon-γ, and chemokines. Moreover, their value as biomarkers in disease development and progression further enhance the validity of this interaction. Recently, cytokine-targeted treatment approaches have emerged as potential tools in the management of atherosclerotic disease. IL-1β, based on the results of the CANTOS trial, remains the most validated option in reducing the residual cardiovascular risk. Along the same line, colchicine was also proven efficacious in preventing major adverse cardiovascular events in large clinical trials of patients with acute and chronic coronary syndrome. Other commercially available agents targeting IL-6 (tocilizumab), TNF-α (etanercept, adalimumab, infliximab), or IL-1 receptor antagonist (anakinra) have mostly been assessed in the setting of other inflammatory diseases and further testing in atherosclerosis is required. In the future, potential targeting of the NLRP3 inflammasome, anti-inflammatory IL-10, or atherogenic chemokines could represent appealing options, provided that patient safety is proven to be of no concern.
Collapse
Affiliation(s)
| | | | | | - Dimitris Tousoulis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, 11527 Athens, Greece
| |
Collapse
|
19
|
Lee SH, Jeong YJ, Park J, Kim HY, Son Y, Kim KS, Lee HJ. Low-Dose Radiation Affects Cardiovascular Disease Risk in Human Aortic Endothelial Cells by Altering Gene Expression under Normal and Diabetic Conditions. Int J Mol Sci 2022; 23:8577. [PMID: 35955709 PMCID: PMC9369411 DOI: 10.3390/ijms23158577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
High doses of ionizing radiation can cause cardiovascular diseases (CVDs); however, the effects of <100 mGy radiation on CVD remain underreported. Endothelial cells (ECs) play major roles in cardiovascular health and disease, and their function is reduced by stimuli such as chronic disease, metabolic disorders, and smoking. However, whether exposure to low-dose radiation results in the disruption of similar molecular mechanisms in ECs under diabetic and non-diabetic states remains largely unknown; we aimed to address this gap in knowledge through the molecular and functional characterization of primary human aortic endothelial cells (HAECs) derived from patients with type 2 diabetes (T2D-HAECs) and normal HAECs in response to low-dose radiation. To address these limitations, we performed RNA sequencing on HAECs and T2D-HAECs following exposure to 100 mGy of ionizing radiation and examined the transcriptome changes associated with the low-dose radiation. Compared with that in the non-irradiation group, low-dose irradiation induced 243 differentially expressed genes (DEGs) (133 down-regulated and 110 up-regulated) in HAECs and 378 DEGs (195 down-regulated and 183 up-regulated) in T2D-HAECs. We also discovered a significant association between the DEGs and the interferon (IFN)-I signaling pathway, which is associated with CVD by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, protein−protein network analysis, and module analysis. Our findings demonstrate the potential impact of low-dose radiation on EC functions that are related to the risk of CVD.
Collapse
Affiliation(s)
- Soo-Ho Lee
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Ye Ji Jeong
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Jeongwoo Park
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Hyun-Yong Kim
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Yeonghoon Son
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Kwang Seok Kim
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| | - Hae-June Lee
- Divisions of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Korea; (S.-H.L.); (Y.J.J.); (J.P.); (H.-Y.K.); (Y.S.)
| |
Collapse
|
20
|
Gu SZ, Bennett MR. Plaque Structural Stress: Detection, Determinants and Role in Atherosclerotic Plaque Rupture and Progression. Front Cardiovasc Med 2022; 9:875413. [PMID: 35872913 PMCID: PMC9300846 DOI: 10.3389/fcvm.2022.875413] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/10/2022] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis remains a major cause of death worldwide, with most myocardial infarctions being due to rupture or erosion of coronary plaques. Although several imaging modalities can identify features that confer risk, major adverse cardiovascular event (MACE) rates attributable to each plaque are low, such that additional biomarkers are required to improve risk stratification at plaque and patient level. Coronary arteries are exposed to continual mechanical forces, and plaque rupture occurs when plaque structural stress (PSS) exceeds its mechanical strength. Prospective studies have shown that peak PSS is correlated with acute coronary syndrome (ACS) presentation, plaque rupture, and MACE, and provides additional prognostic information to imaging. In addition, PSS incorporates multiple variables, including plaque architecture, plaque material properties, and haemodynamic data into a defined solution, providing a more detailed overview of higher-risk lesions. We review the methods for calculation and determinants of PSS, imaging modalities used for modeling PSS, and idealized models that explore structural and geometric components that affect PSS. We also discuss current experimental and clinical data linking PSS to the natural history of coronary artery disease, and explore potential for refining treatment options and predicting future events.
Collapse
Affiliation(s)
| | - Martin R. Bennett
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
21
|
Khare HA, Døssing KBV, Ringgaard L, Christensen E, Urbak L, Sillesen H, Ripa RS, Binderup T, Pedersen SF, Kjaer A. In vivo detection of urokinase-type plasminogen activator receptor (uPAR) expression in arterial atherogenesis using [64Cu]Cu-DOTA-AE105 positron emission tomography (PET). Atherosclerosis 2022; 352:103-111. [DOI: 10.1016/j.atherosclerosis.2022.03.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/23/2022] [Accepted: 03/25/2022] [Indexed: 12/21/2022]
|
22
|
Pallarès J, Torreguitart N, Arqué G, Portero-Otin M, Purroy F. Human atheromatous plaques expressed sensing adaptor STING, a potential role in vascular inflammation pathogenesis. Thromb Haemost 2022; 122:1621-1624. [PMID: 35170007 DOI: 10.1055/a-1772-1192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Atherogenesis is a complex physiological process involving inflammation and contributing to plaque development and vulnerability. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has emerged as a significant intracellular mechanism of innate immune defense programs. However, its involvement in atherogenesis has not been explored. In this brief report, we assessed the expression of STING protein in the artheriosclerotic (ATC) plaque of human patients. Our results showed that STING expression was detected in the ATC plaques. Most of the STING expression was noted in macrophages and lymphocytes. Of note, endothelial cells were constantly positive in all cases analyzed. STING expression was strong in the complicated ATC plaques near the cell debris and hemorrhagic foci. Considering these findings, we propose that cGAS-STING signaling plays a role in atherogenesis, opening novel avenues for therapeutic development.
Collapse
Affiliation(s)
- Judit Pallarès
- Pahology, Arnau de Vilanova University Hospital, Lleida, Spain
| | | | | | | | | |
Collapse
|
23
|
Nettersheim FS, Picard FSR, Hoyer FF, Winkels H. Immunotherapeutic Strategies in Cancer and Atherosclerosis-Two Sides of the Same Coin. Front Cardiovasc Med 2022; 8:812702. [PMID: 35097027 PMCID: PMC8792753 DOI: 10.3389/fcvm.2021.812702] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Friedrich Felix Hoyer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Rohde D, Vandoorne K, Lee IH, Grune J, Zhang S, McAlpine CS, Schloss MJ, Nayar R, Courties G, Frodermann V, Wojtkiewicz G, Honold L, Chen Q, Schmidt S, Iwamoto Y, Sun Y, Cremer S, Hoyer FF, Iborra-Egea O, Muñoz-Guijosa C, Ji F, Zhou B, Adams RH, Wythe JD, Hidalgo J, Watanabe H, Jung Y, van der Laan AM, Piek JJ, Kfoury Y, Désogère PA, Vinegoni C, Dutta P, Sadreyev RI, Caravan P, Bayes-Genis A, Libby P, Scadden DT, Lin CP, Naxerova K, Swirski FK, Nahrendorf M. Bone marrow endothelial dysfunction promotes myeloid cell expansion in cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:28-44. [PMID: 35747128 PMCID: PMC9216333 DOI: 10.1038/s44161-021-00002-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/27/2021] [Indexed: 12/13/2022]
Abstract
Abnormal hematopoiesis advances cardiovascular disease by generating excess inflammatory leukocytes that attack the arteries and the heart. The bone marrow niche regulates hematopoietic stem cell proliferation and hence the systemic leukocyte pool, but whether cardiovascular disease affects the hematopoietic organ's microvasculature is unknown. Here we show that hypertension, atherosclerosis and myocardial infarction (MI) instigate endothelial dysfunction, leakage, vascular fibrosis and angiogenesis in the bone marrow, altogether leading to overproduction of inflammatory myeloid cells and systemic leukocytosis. Limiting angiogenesis with endothelial deletion of Vegfr2 (encoding vascular endothelial growth factor (VEGF) receptor 2) curbed emergency hematopoiesis after MI. We noted that bone marrow endothelial cells assumed inflammatory transcriptional phenotypes in all examined stages of cardiovascular disease. Endothelial deletion of Il6 or Vcan (encoding versican), genes shown to be highly expressed in mice with atherosclerosis or MI, reduced hematopoiesis and systemic myeloid cell numbers in these conditions. Our findings establish that cardiovascular disease remodels the vascular bone marrow niche, stimulating hematopoiesis and production of inflammatory leukocytes.
Collapse
Affiliation(s)
- David Rohde
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Cardiology, Angiology and Pneumology, Heidelberg University Hospital, Heidelberg, Germany
- These authors contributed equally: David Rohde, Katrien Vandoorne
| | - Katrien Vandoorne
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Biomedical Engineering Faculty, Technion-Israel Institute of Technology, Haifa, Israel
- These authors contributed equally: David Rohde, Katrien Vandoorne
| | - I-Hsiu Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jana Grune
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuang Zhang
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Cameron S. McAlpine
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maximilian J. Schloss
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ribhu Nayar
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabriel Courties
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Vanessa Frodermann
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lisa Honold
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Qi Chen
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Stephen Schmidt
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yuan Sun
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Sebastian Cremer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Friedrich F. Hoyer
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Fei Ji
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Bin Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Ralf H. Adams
- Max Planck Institute for Molecular Biomedicine, Muenster, Germany
| | - Joshua D. Wythe
- Cardiovascular Research Institute, Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Juan Hidalgo
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi, Japan
| | - Yookyung Jung
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anja M. van der Laan
- Heart Center, Department of Cardiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan J. Piek
- Heart Center, Department of Cardiology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Youmna Kfoury
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Pauline A. Désogère
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Partha Dutta
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Caravan
- Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | | | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - David T. Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Charles P. Lin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Institute for Molecular Science of Medicine, Aichi Medical University, Aichi, Japan
| | - Kamila Naxerova
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Filip K. Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
25
|
Sallam M, Benotmane MA, Baatout S, Guns PJ, Aerts A. Radiation-induced cardiovascular disease: an overlooked role for DNA methylation? Epigenetics 2022; 17:59-80. [PMID: 33522387 PMCID: PMC8812767 DOI: 10.1080/15592294.2021.1873628] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/27/2020] [Accepted: 01/04/2021] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy in cancer treatment involves the use of ionizing radiation for cancer cell killing. Although radiotherapy has shown significant improvements on cancer recurrence and mortality, several radiation-induced adverse effects have been documented. Of these adverse effects, radiation-induced cardiovascular disease (CVD) is particularly prominent among patients receiving mediastinal radiotherapy, such as breast cancer and Hodgkin's lymphoma patients. A number of mechanisms of radiation-induced CVD pathogenesis have been proposed such as endothelial inflammatory activation, premature endothelial senescence, increased ROS and mitochondrial dysfunction. However, current research seems to point to a so-far unexamined and potentially novel involvement of epigenetics in radiation-induced CVD pathogenesis. Firstly, epigenetic mechanisms have been implicated in CVD pathophysiology. In addition, several studies have shown that ionizing radiation can cause epigenetic modifications, especially DNA methylation alterations. As a result, this review aims to provide a summary of the current literature linking DNA methylation to radiation-induced CVD and thereby explore DNA methylation as a possible contributor to radiation-induced CVD pathogenesis.
Collapse
Affiliation(s)
- Magy Sallam
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - Mohammed Abderrafi Benotmane
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, University of Antwerp, Wilrijk, Belgium
| | - An Aerts
- Radiobiology Unit, Institute for Environment, Health and Safety, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| |
Collapse
|
26
|
Iwanski J, Kazmouz SG, Li S, Stansfield B, Salem TT, Perez-Miller S, Kazui T, Jena L, Uhrlaub JL, Lick S, Nikolich-Žugich J, Konhilas JP, Gregorio CC, Khanna M, Campos SK, Churko JM. Antihypertensive drug treatment and susceptibility to SARS-CoV-2 infection in human PSC-derived cardiomyocytes and primary endothelial cells. Stem Cell Reports 2021; 16:2459-2472. [PMID: 34525378 PMCID: PMC8407952 DOI: 10.1016/j.stemcr.2021.08.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 01/08/2023] Open
Abstract
The pathogenicity of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been attributed to its ability to enter through the membrane-bound angiotensin-converting enzyme 2 (ACE2) receptor. Therefore, it has been heavily speculated that angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor blocker (ARB) therapy may modulate SARS-CoV-2 infection. In this study, exposure of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and human endothelial cells (hECs) to SARS-CoV-2 identified significant differences in protein coding genes involved in immunity, viral response, and cardiomyocyte/endothelial structure. Specifically, transcriptome changes were identified in the tumor necrosis factor (TNF), interferon α/β, and mitogen-activated protein kinase (MAPK) (hPSC-CMs) as well as nuclear factor kappa-B (NF-κB) (hECs) signaling pathways. However, pre-treatment of hPSC-CMs or hECs with two widely prescribed antihypertensive medications, losartan and lisinopril, did not affect the susceptibility of either cell type to SARS-CoV-2 infection. These findings demonstrate the toxic effects of SARS-CoV-2 in hPSC-CMs/hECs and, taken together with newly emerging multicenter trials, suggest that antihypertensive drug treatment alone does not alter SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jessika Iwanski
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Sobhi G Kazmouz
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Shuaizhi Li
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA
| | - Ben Stansfield
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA; Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Tori T Salem
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA; Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Toshinobu Kazui
- Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| | - Lipsa Jena
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA
| | - Scott Lick
- Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA; BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| | - John P Konhilas
- Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Samuel K Campos
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA; Department of Molecular & Cellular Biology, The University of Arizona, Tucson, AZ, USA; Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA; BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| | - Jared M Churko
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA; Department of Physiology, The University of Arizona, Tucson, AZ, USA; BIO5 Institute, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
27
|
Parra-Izquierdo I, Sánchez-Bayuela T, López J, Gómez C, Pérez-Riesgo E, San Román JA, Sánchez Crespo M, Yacoub M, Chester AH, García-Rodríguez C. Interferons Are Pro-Inflammatory Cytokines in Sheared-Stressed Human Aortic Valve Endothelial Cells. Int J Mol Sci 2021; 22:ijms221910605. [PMID: 34638942 PMCID: PMC8508640 DOI: 10.3390/ijms221910605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve disease (CAVD) is an athero-inflammatory process. Growing evidence supports the inflammation-driven calcification model, mediated by cytokines such as interferons (IFNs) and tumor necrosis factor (TNF)-α. Our goal was investigating IFNs' effects in human aortic valve endothelial cells (VEC) and the potential differences between aortic (aVEC) and ventricular (vVEC) side cells. The endothelial phenotype was analyzed by Western blot, qPCR, ELISA, monocyte adhesion, and migration assays. In mixed VEC populations, IFNs promoted the activation of signal transducers and activators of transcription-1 and nuclear factor-κB, and the subsequent up-regulation of pro-inflammatory molecules. Side-specific VEC were activated with IFN-γ and TNF-α in an orbital shaker flow system. TNF-α, but not IFN-γ, induced hypoxia-inducible factor (HIF)-1α stabilization or endothelial nitric oxide synthase downregulation. Additionally, IFN-γ inhibited TNF-α-induced migration of aVEC. Also, IFN-γ triggered cytokine secretion and adhesion molecule expression in aVEC and vVEC. Finally, aVEC were more prone to cytokine-mediated monocyte adhesion under multiaxial flow conditions as compared with uniaxial flow. In conclusion, IFNs promote inflammation and reduce TNF-α-mediated migration in human VEC. Moreover, monocyte adhesion was higher in inflamed aVEC sheared under multiaxial flow, which may be relevant to understanding the initial stages of CAVD.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Tania Sánchez-Bayuela
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Javier López
- ICICOR, Hospital Clínico Universitario, 47005 Valladolid, Spain; (J.L.); (J.A.S.R.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Cristina Gómez
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Enrique Pérez-Riesgo
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - J. Alberto San Román
- ICICOR, Hospital Clínico Universitario, 47005 Valladolid, Spain; (J.L.); (J.A.S.R.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
| | - Magdi Yacoub
- National Heart & Lung Institute, Imperial College London, London SW3 6LR, UK;
- Magdi Yacoub Institute, Harefield UB9 6JH, UK
| | - Adrian H. Chester
- National Heart & Lung Institute, Imperial College London, London SW3 6LR, UK;
- Magdi Yacoub Institute, Harefield UB9 6JH, UK
- Correspondence: (A.H.C.); (C.G.-R.); Tel.: +44-(0)1895-760732 (A.H.C.); +34-983-184841 (C.G.-R.)
| | - Carmen García-Rodríguez
- Instituto de Biología y Genética Molecular, Spanish National Research Council (CSIC), Universidad de Valladolid, 47003 Valladolid, Spain; (I.P.-I.); (T.S.-B.); (C.G.); (E.P.-R.); (M.S.C.)
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence: (A.H.C.); (C.G.-R.); Tel.: +44-(0)1895-760732 (A.H.C.); +34-983-184841 (C.G.-R.)
| |
Collapse
|
28
|
Ballasy NN, Jadli AS, Edalat P, Kang S, Fatehi Hassanabad A, Gomes KP, Fedak PWM, Patel VB. Potential role of epicardial adipose tissue in coronary artery endothelial cell dysfunction in type 2 diabetes. FASEB J 2021; 35:e21878. [PMID: 34469050 DOI: 10.1096/fj.202100684rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 11/11/2022]
Abstract
Cardiovascular disease is the most prevalent cause of morbidity and mortality in diabetes. Epicardial adipose tissue (EAT) lies in direct contact with the myocardium and coronary arteries and can influence cardiac (patho) physiology through paracrine signaling pathways. This study hypothesized that the proteins released from EAT represent a critical molecular link between the diabetic state and coronary artery endothelial cell dysfunction. To simulate type 2 diabetes-associated metabolic and inflammatory status in an ex vivo tissue culture model, human EAT samples were treated with a cocktail composed of high glucose, high palmitate, and lipopolysaccharide (gplEAT) and were compared with control EAT (conEAT). Compared to conEAT, gplEAT showed a markedly increased gene expression profile of proinflammatory cytokines, corroborating EAT inflammation, a hallmark feature observed in patients with type 2 diabetes. Luminex assay of EAT-secretome identified increased release of various proinflammatory cytokines, including tumor necrosis factor-alpha (TNF-alpha), interferon-alpha 2 (IFNA2), interleukin 1 beta (IL1B), interleukin 5 (IL5), interleukin 13 (IL13), and CCL5, among others, in response to high glucose, high palmitate, and lipopolysaccharide. Conditioned culture media was used to collect the concentrated proteins (CPs). In response to gplEAT-CPs, human coronary artery endothelial cells (HCAECs) exhibited an inflammatory endothelial cell phenotype, featuring a significantly increased gene expression of proinflammatory cytokines and cell surface expression of VCAM-1. Moreover, gplEAT-CPs severely decreased Akt-eNOS signaling, nitric oxide production, and angiogenic potential of HCAECs, when compared with conEAT-CPs. These findings indicate that EAT inflammation may play a key role in coronary artery endothelial cell dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- Noura N Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Anshul S Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Pariya Edalat
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Sean Kang
- Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.,Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ali Fatehi Hassanabad
- Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.,Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Karina P Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| | - Paul W M Fedak
- Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada.,Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vaibhav B Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Libin Cardiovascular Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
29
|
Zafar A, Pong Ng H, Diamond-Zaluski R, Kim GD, Ricky Chan E, Dunwoodie SL, Smith JD, Mahabeleshwar GH. CITED2 inhibits STAT1-IRF1 signaling and atherogenesis. FASEB J 2021; 35:e21833. [PMID: 34365659 DOI: 10.1096/fj.202100792r] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 11/11/2022]
Abstract
Macrophages are the principal component of the innate immune system. They play very crucial and multifaceted roles in the pathogenesis of inflammatory vascular diseases. There is an increasing recognition that transcriptionally dynamic macrophages are the key players in the pathogenesis of inflammatory vascular diseases. In this context, the accumulation and aberrant activation of macrophages in the subendothelial layers govern atherosclerotic plaque development. Macrophage-mediated inflammation is an explicitly robust biological response that involves broad alterations in inflammatory gene expression. Thus, cell-intrinsic negative regulatory mechanisms must exist which can restrain inflammatory response in a spatiotemporal manner. In this study, we identified CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) as one such cell-intrinsic negative regulator of inflammation. Our in vivo studies show that myeloid-CITED2-deficient mice on the Apoe-/- background have larger atherosclerotic lesions on both control and high-fat/high-cholesterol diets. Our integrated transcriptomics and gene set enrichment analyses studies show that CITED2 deficiency elevates STAT1 and interferon regulatory factor 1 (IRF1) regulated pro-inflammatory gene expression in macrophages. At the molecular level, our studies identify that CITED2 deficiency elevates IFNγ-induced STAT1 transcriptional activity and STAT1 enrichment on IRF1 promoter in macrophages. More importantly, siRNA-mediated knockdown of IRF1 completely reversed elevated pro-inflammatory target gene expression in CITED2-deficient macrophages. Collectively, our study findings demonstrate that CITED2 restrains the STAT1-IRF1 signaling axis in macrophages and limits the development of atherosclerotic plaques.
Collapse
Affiliation(s)
- Atif Zafar
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Hang Pong Ng
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rachel Diamond-Zaluski
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Gun-Dong Kim
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ernest Ricky Chan
- Cleveland Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sally L Dunwoodie
- Victor Chang Cardiac Research Institute, Sydney, NSW, Australia.,Faculties of Medicine and Science, UNSW Sydney, Sydney, NSW, Australia
| | - Jonathan D Smith
- Department of Cardiovascular & Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
30
|
Baldini C, Moriconi FR, Galimberti S, Libby P, De Caterina R. The JAK-STAT pathway: an emerging target for cardiovascular disease in rheumatoid arthritis and myeloproliferative neoplasms. Eur Heart J 2021; 42:4389-4400. [PMID: 34343257 DOI: 10.1093/eurheartj/ehab447] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/21/2021] [Accepted: 07/31/2021] [Indexed: 01/07/2023] Open
Abstract
Inflammation contributes centrally to cardiovascular diseases, and anti-inflammatory treatments can reduce cardiovascular events. The JAK-STAT pathway is an emerging target in inflammation, mainly in rheumatoid arthritis (RA) and chronic myeloproliferative neoplasms (MPNs), disorders that heighten cardiovascular risk. The aim of this study was to review the international literature on the relationship between dysregulation of the JAK-STAT pathway in RA/MPNs and cardiovascular risk and on the potential cardiovascular effects of JAK-STAT inhibitors. The JAK-STAT pathway sustains inflammatory and thrombotic events in autoimmune disorders such as RA and MPNs. Here, an imbalance exists between pro- and anti-inflammatory cytokines [increased levels of interleukin (IL)-6, IL-1-β, tumour necrosis factor-α, decreased levels of IL-10] and the over-expression of some prothrombotic proteins, such as protein kinase Cε, on the surface of activated platelets. This pathway also operates in atherosclerotic cardiovascular disease. JAK-STAT inhibitors may reduce cardiovascular events and related deaths in such conditions, but the potential of these agents requires more studies, especially with regard to cardiovascular safety, and particularly for potential prothrombotic effects. JAK-STAT inhibitors merit consideration to curb heightened cardiovascular risk in patients with RA and MPNs, with rigorous assessment of the potential benefits and risks.
Collapse
Affiliation(s)
- Chiara Baldini
- Division of Rheumatology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| | - Francesca Romana Moriconi
- Division of Rheumatology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy.,Division of Cardiology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| | - Sara Galimberti
- Division of Hematology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| | - Peter Libby
- Cardiovascular Division, Brigham and Women's Hospital-Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Raffaele De Caterina
- Division of Cardiology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| |
Collapse
|
31
|
Łuczak A, Małecki R, Kulus M, Madej M, Szahidewicz-Krupska E, Doroszko A. Cardiovascular Risk and Endothelial Dysfunction in Primary Sjogren Syndrome Is Related to the Disease Activity. Nutrients 2021; 13:nu13062072. [PMID: 34204342 PMCID: PMC8235705 DOI: 10.3390/nu13062072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/14/2021] [Accepted: 06/14/2021] [Indexed: 12/23/2022] Open
Abstract
The aim of our study was to evaluate if endothelial-dysfunction (ED) occurs in patients with primary Sjogren syndrome (pSS) and whether it is associated with the disease characteristics and activity. A total of 46 patients with pSS and 30 controls, without known cardiovascular disease, were enrolled in this study. A flow-mediated-dilation (FMD) of the brachial artery, plasma concentrations of the nitric oxide (NO) metabolic pathway (ADMA, L-arginine, SDMA, cGMP), and markers of endothelial inflammatory function (PAI-1, sE-selectin) and angiogenesis (angiostatin, VEGF) were analyzed. The FMD was significantly lower in pSS patients (7.56 ± 3.08 vs. 10.91 ± 1.02%, p = 0.043) and positively correlated with the Ro/SS-A-antibodies (r = 0.34, p = 0.03), pulmonary involvement (r = 0.52, p = 0.001) and inversely with ADMA (r = −0.35, p = 0.04). Plasma ADMA, L-arginine and angiostatin levels were significantly higher in pSS patients (0.39 ± 0.08 vs. 0.36 ± 0.06 µmol/L, p = 0.05; 29.07 ± 6.7 vs. 25.4 ± 5.23 µmol/L, p = 0.01; 152.25 ± 60.99 vs. 120.07 ± 38.7 pg/mL, p = 0.0, respectively). ADMA was associated with ESSDAI (r = 0.33, p = 0.02), SCORE (r = 0.57, p = 0.00003) and focus score (r = 0.38, p = 0.04). In the multiple regression analysis, the ESSDAI was significantly and independently associated with plasma ADMA levels (β = 0.24, p = 0.04). Moreover, plasma cGMP concentrations were negatively correlated with the disease duration (r = −0.31, p = 0.03). Endothelial function is impaired in patients with pSS and associated with the measures of disease activity, which supports the key-role of inflammation in developing and maintaining accelerated atherosclerosis.
Collapse
Affiliation(s)
- Anna Łuczak
- Department of Rheumatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (A.Ł.); (M.M.)
| | - Rafał Małecki
- Department of Angiology, Hypertension and Diabetology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Michał Kulus
- Department of Histology and Embryology, Wroclaw Medical University, Chalubinskiego 6a, 50-367 Wroclaw, Poland;
| | - Marta Madej
- Department of Rheumatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland; (A.Ł.); (M.M.)
| | - Ewa Szahidewicz-Krupska
- Department of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Adrian Doroszko
- Department of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-71-736-4000
| |
Collapse
|
32
|
Santinelli L, De Girolamo G, Borrazzo C, Vassalini P, Pinacchio C, Cavallari EN, Statzu M, Frasca F, Scordio M, Bitossi C, Viscido A, Ceccarelli G, Mancone M, Mastroianni CM, Antonelli G, d'Ettorre G, Scagnolari C. Alteration of type I interferon response is associated with subclinical atherosclerosis in virologically suppressed HIV-1-infected male patients. J Med Virol 2021; 93:4930-4938. [PMID: 33913525 PMCID: PMC8360015 DOI: 10.1002/jmv.27028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
Given human immunodeficiency virus‐1 (HIV‐1)‐infected patients have alterations in the type I interferon (IFN‐I) pathway and are also at elevated risk of atherosclerosis, we evaluated IFN‐I response and subclinical cardiovascular disease (CVD) association in HIV‐1‐infected patients. Transcript levels of IFN‐α/β and IFN‐stimulated gene 56 (ISG56) were evaluated by RT/real‐time PCR in peripheral blood mononuclear cells collected from asymptomatic HIV‐1‐positive male patients at high risk of developing CVD (n = 34) and healthy subjects (n = 21). Stenosis degree (≥ or <50%), calcium volume score, calcium Agatston score, and myocardial extracellular volume were examined by coronary computerized tomography scan. Carotid intima‐media thickness (cIMT), Framingham risk score, atherosclerotic cardiovascular disease (ASCVD) score, and risk score developed by data collection on adverse effects of anti‐HIV drugs (D:A:D) were also measured. Increased IFN‐α, IFN‐β, and ISG56 levels were observed in all HIV‐1‐infected males compared to healthy controls (p < .001 for all genes analyzed). HIV‐1‐infected patients with a stenosis degree ≥50% showed a higher Framingham risk score (p = .019), which was correlated with IFN‐β and ISG56 levels. HIV‐1‐infected males with enhanced IFN‐I levels and stenosis displayed a higher ASCVD calculated risk (p = .011) and D:A:D score (p = .004). Also, there was a trend toward higher IFN‐α and ISG56 mRNA levels in HIV‐1‐positive patients with an increased cIMT (p > .05). Dysregulation of IFN‐I response might participate in the pathogenesis of HIV‐1‐associated CVD.
Collapse
Affiliation(s)
- Letizia Santinelli
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| | - Gabriella De Girolamo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Cristian Borrazzo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Paolo Vassalini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Claudia Pinacchio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Eugenio Nelson Cavallari
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Maura Statzu
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| | - Federica Frasca
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| | - Mirko Scordio
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| | - Camilla Bitossi
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| | - Agnese Viscido
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Massimo Mancone
- Department of Cardiovascular, Respiratory, Nephrology, Anaesthesiology and Geriatric Sciences, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Claudio Maria Mastroianni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Guido Antonelli
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy.,Microbiology and Virology Unit, Sapienza University Hospital "Policlinico Umberto I", Rome, Italy
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Viale di Porta Tiburtina, Rome, Italy
| |
Collapse
|
33
|
Afonso MS, Sharma M, Schlegel M, van Solingen C, Koelwyn GJ, Shanley LC, Beckett L, Peled D, Rahman K, Giannarelli C, Li H, Brown EJ, Khodadadi-Jamayran A, Fisher EA, Moore KJ. miR-33 Silencing Reprograms the Immune Cell Landscape in Atherosclerotic Plaques. Circ Res 2021; 128:1122-1138. [PMID: 33593073 PMCID: PMC8049965 DOI: 10.1161/circresaha.120.317914] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/15/2021] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Milessa Silva Afonso
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Monika Sharma
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Martin Schlegel
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
- Department of Anesthesiology and Intensive Care, Technical University of Munich School of Medicine, Germany (M. Schlegel)
| | - Coen van Solingen
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Graeme J Koelwyn
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Lianne C Shanley
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Lauren Beckett
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
| | - Daniel Peled
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Karishma Rahman
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Chiara Giannarelli
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY (C.G.)
| | - Huilin Li
- Division of Biostatics, Department of Population Health (H.L), New York University School of Medicine
| | - Emily J Brown
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | | | - Edward A Fisher
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| | - Kathryn J Moore
- Department of Medicine, Leon H. Charney Division of Cardiology, New York University Grossman School of Medicine (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.)
- NYU Cardiovascular Research Center (M.S.A., M. Sharma, M. Schlegel, C.v.S., G.J.K., L.C.S., L.B., D.P., K.R., E.J.B., E.A.F., K.J.M.), New York University School of Medicine
| |
Collapse
|
34
|
Li YQ, Li YL, Li XT, Lv JY, Gao Y, Li WN, Gong QH, Yang DL. Osthole Alleviates Neointimal Hyperplasia in Balloon-Induced Arterial Wall Injury by Suppressing Vascular Smooth Muscle Cell Proliferation and Downregulating Cyclin D1/CDK4 and Cyclin E1/CDK2 Expression. Front Physiol 2021; 11:514494. [PMID: 33574763 PMCID: PMC7870719 DOI: 10.3389/fphys.2020.514494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/30/2020] [Indexed: 11/13/2022] Open
Abstract
Percutaneous coronary intervention (PCI) is the most widely used therapy for treating ischemic heart disease. However, intimal hyperplasia and restenosis usually occur within months after angioplasty. Modern pharmacological researchers have proven that osthole, the major active coumarin of Cnidium monnieri (L.) Cusson, exerts potent antiproliferative effects in lung cancer cells, the human laryngeal cancer cell line RK33 and TE671 medulloblastoma cells, and its mechanism of action is related to cell cycle arrest. The goal of the present study was to observe the effect of osthole on vascular smooth muscle cell (VSMC) proliferation using platelet-derived growth factor-BB (PDGF-BB)-stimulated VSMCs isolated from rats and vascular balloon injury as models to further elucidate the molecular mechanisms underlying this activity. We detected the relative number of VSMCs by the MTT assay and EdU staining and examined cell cycle progression by flow cytometry. To more deeply probe the mechanisms, the protein expression levels of PCNA, the cyclin D1/CDK4 complex and the cyclin E1/CDK2 complex in balloon-treated rat carotid arteries and the mRNA and protein expression levels of the cyclin D1/CDK4 and cyclin E1/CDK2 complexes in VSMCs were detected by real-time RT-PCR and western blotting. The data showed that osthole significantly inhibited the proliferation of VSMCs induced by PDGF-BB. Furthermore, osthole caused apparent VSMC cycle arrest early in G0/G1 phase and decreased the expression of cyclin D1/CDK4 and cyclin E1/CDK2. Our results demonstrate that osthole can significantly inhibit PDGF-BB-induced VSMC proliferation and that its regulatory effects on cell cycle progression and proliferation may be related to the downregulation of cyclin D1/CDK4 and cyclin E1/CDK2 expression as well as the prevention of cell cycle progression from G0/G1 phase to S phase. The abovementioned mechanism may be responsible for the alleviation of neointimal hyperplasia in balloon-induced arterial wall injury by osthole.
Collapse
Affiliation(s)
- Yi-Qi Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of the Ministry of Education, The Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Department of Pharmacology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Ye-Li Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of the Ministry of Education, The Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xiao-Tong Li
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of the Ministry of Education, The Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jun-Yuan Lv
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yang Gao
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of the Ministry of Education, The Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Wen-Na Li
- Department of Pharmacology, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Qi-Hai Gong
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of the Ministry of Education, The Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Dan-Li Yang
- Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of the Ministry of Education, The Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, School of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
35
|
Kwiatkowska I, Hermanowicz JM, Mysliwiec M, Pawlak D. Oxidative Storm Induced by Tryptophan Metabolites: Missing Link between Atherosclerosis and Chronic Kidney Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6656033. [PMID: 33456671 PMCID: PMC7787774 DOI: 10.1155/2020/6656033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023]
Abstract
Chronic kidney disease (CKD) occurrence is rising all over the world. Its presence is associated with an increased risk of premature death from cardiovascular disease (CVD). Several explanations of this link have been put forward. It is known that in renal failure, an array of metabolites cannot be excreted, and they accumulate in the organism. Among them, some are metabolites of tryptophan (TRP), such as indoxyl sulfate and kynurenine. Scientists have become interested in them in the context of inducing vascular damage in the course of chronic kidney impairment. Experimental evidence suggests the involvement of TRP metabolites in the progression of chronic kidney disease and atherosclerosis separately and point to oxidative stress generation as one of the main mechanisms that is responsible for worsening those states. Since it is known that blood levels of those metabolites increase significantly in renal failure and that they generate reactive oxygen species (ROS), which lead to endothelial injury, it is reasonable to suspect that products of TRP metabolism are the missing link in frequently occurring atherosclerosis in CKD patients. This review focuses on reports that shed a light on TRP metabolites as contributing factors to vascular damage in the progression of impaired kidney function.
Collapse
Affiliation(s)
- Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Justyna M. Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Michal Mysliwiec
- Ist Department Nephrology and Transplantation, Medical University, Bialystok, Zurawia 14, 15-540 Bialystok, Poland
- Lomza State University of Applied Sciences, Akademicka 14, 18-400 Łomża, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
- Department of Pharmacology and Toxicology, University of Warmia and Mazury in Olsztyn, Warszawska 30, 10-082 Olsztyn, Poland
| |
Collapse
|
36
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Wasiak S, Dzobo KE, Rakai BD, Kaiser Y, Versloot M, Bahjat M, Stotz SC, Fu L, Sweeney M, Johansson JO, Wong NCW, Stroes ESG, Kroon J, Kulikowski E. BET protein inhibitor apabetalone (RVX-208) suppresses pro-inflammatory hyper-activation of monocytes from patients with cardiovascular disease and type 2 diabetes. Clin Epigenetics 2020; 12:166. [PMID: 33172487 PMCID: PMC7657365 DOI: 10.1186/s13148-020-00943-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Patients with cardiovascular disease (CVD) and type 2 diabetes (DM2) have a high residual risk for experiencing a major adverse cardiac event. Dysregulation of epigenetic mechanisms of gene transcription in innate immune cells contributes to CVD development but is currently not targeted by therapies. Apabetalone (RVX-208) is a small molecule inhibitor of bromodomain and extra-terminal (BET) proteins—histone acetylation readers that drive pro-inflammatory and pro-atherosclerotic gene transcription. Here, we assess the impact of apabetalone on ex vivo inflammatory responses of monocytes from DM2 + CVD patients. Results Monocytes isolated from DM2 + CVD patients and matched controls were treated ex vivo with apabetalone, interferon γ (IFNγ), IFNγ + apabetalone or vehicle and phenotyped for gene expression and protein secretion. Unstimulated DM2 + CVD monocytes had higher baseline IL-1α, IL-1β and IL-8 cytokine gene expression and Toll-like receptor (TLR) 2 surface abundance than control monocytes, indicating pro-inflammatory activation. Further, DM2 + CVD monocytes were hyper-responsive to stimulation with IFNγ, upregulating genes within cytokine and NF-κB pathways > 30% more than control monocytes (p < 0.05). Ex vivo apabetalone treatment countered cytokine secretion by DM2 + CVD monocytes at baseline (GROα and IL-8) and during IFNγ stimulation (IL-1β and TNFα). Apabetalone abolished pro-inflammatory hyper-activation by reducing TLR and cytokine gene signatures more robustly in DM2 + CVD versus control monocytes. Conclusions Monocytes isolated from DM2 + CVD patients receiving standard of care therapies are in a hyper-inflammatory state and hyperactive upon IFNγ stimulation. Apabetalone treatment diminishes this pro-inflammatory phenotype, providing mechanistic insight into how BET protein inhibition may reduce CVD risk in DM2 patients.
Collapse
Affiliation(s)
- Sylwia Wasiak
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Kim E Dzobo
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Brooke D Rakai
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Miranda Versloot
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Mahnoush Bahjat
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Stephanie C Stotz
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Li Fu
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Michael Sweeney
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Jan O Johansson
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Norman C W Wong
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ewelina Kulikowski
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada.
| |
Collapse
|
38
|
Dong M, Zhang Y, Xu C, Wang C, Liu M, Zhang Z, Wu H, Yuan Z, Zhou J. Interferon-γ decreases ATP-binding cassette subfamily G member 1-mediated cholesterol efflux through small ubiquitin-like modifier/ubiquitin-dependent liver X receptor-α degradation in macrophages. Biotechnol Appl Biochem 2020; 68:1412-1420. [PMID: 33125792 DOI: 10.1002/bab.2063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 10/24/2020] [Indexed: 11/05/2022]
Abstract
The effects of interferon-γ (IFN-γ) on cholesterol accumulation and the development of foam cells are still unclear. In the present study, we found that IFN-γ promoted liver X receptor (LXR)-α degradation through the ubiquitin-proteasome system in macrophages. The process was dependent on its interactions with phosphorylated signal transducer and activator of transcription 1 (p-STAT1) and protein inhibitor of activated STAT 1 (PIAS1) because both fludarabine and PIAS1 shRNA reversed the decrease in LXR-α protein expression induced by IFN-γ. Additionally, IFN-γ enhanced the interactions of ubiquitin-conjugating enzyme 9 (UBC9), small ubiquitin-like modifier (SUMO)-1 and SUMO-2/3 with LXR-α. Moreover, treatment with shRNA specific for them not only reduced LXR-α polyubiquitination but also reversed the IFN-γ-induced decrease in its expression. Two specific sumoylation sites in LXR-α, K22 and K326, were indispensable for its IFN-γ-induced polyubiquitination because the K22R and K326R mutations inhibited the polyubiquitination and degradation of LXR-α in IFN-γ-treated macrophages. In addition, K22R or K326R mutation almost completely restored ATP-binding cassette subfamily G member 1 (ABCG1)-mediated cholesterol efflux in IFN-γ-treated macrophages. Taken together, these findings indicate that IFN-γ promotes LXR-α degradation through a SUMO-ubiquitin-dependent pathway, which may inhibit cholesterol efflux mediated by ABCG1 from macrophages and promote the development of atherosclerosis.
Collapse
Affiliation(s)
- Mengya Dong
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Yan Zhang
- Department of Cardiovascular Medicine, Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chenbo Xu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chen Wang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Mengping Liu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Zhanyi Zhang
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Haoyu Wu
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Juan Zhou
- Department of Cardiovascular Medicine, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,Key Laboratory of Molecular Cardiology of Shaanxi Province, Xi'an, People's Republic of China
| |
Collapse
|
39
|
Gopal R, Marinelli MA, Alcorn JF. Immune Mechanisms in Cardiovascular Diseases Associated With Viral Infection. Front Immunol 2020; 11:570681. [PMID: 33193350 PMCID: PMC7642610 DOI: 10.3389/fimmu.2020.570681] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
Influenza virus infection causes 3-5 million cases of severe illness and 250,000-500,000 deaths worldwide annually. Although pneumonia is the most common complication associated with influenza, there are several reports demonstrating increased risk for cardiovascular diseases. Several clinical case reports, as well as both prospective and retrospective studies, have shown that influenza can trigger cardiovascular events including myocardial infarction (MI), myocarditis, ventricular arrhythmia, and heart failure. A recent study has demonstrated that influenza-infected patients are at highest risk of having MI during the first seven days of diagnosis. Influenza virus infection induces a variety of pro-inflammatory cytokines and chemokines and recruitment of immune cells as part of the host immune response. Understanding the cellular and molecular mechanisms involved in influenza-associated cardiovascular diseases will help to improve treatment plans. This review discusses the direct and indirect effects of influenza virus infection on triggering cardiovascular events. Further, we discussed the similarities and differences in epidemiological and pathogenic mechanisms involved in cardiovascular events associated with coronavirus disease 2019 (COVID-19) compared to influenza infection.
Collapse
Affiliation(s)
- Radha Gopal
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States
| | | | | |
Collapse
|
40
|
Elyasi A, Voloshyna I, Ahmed S, Kasselman LJ, Behbodikhah J, De Leon J, Reiss AB. The role of interferon-γ in cardiovascular disease: an update. Inflamm Res 2020; 69:975-988. [PMID: 32699989 DOI: 10.1007/s00011-020-01382-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is the leading cause of death, globally, and its prevalence is only expected to rise due to the increasing incidence of co-morbidities such as obesity and diabetes. Medical treatment of CVD is directed primarily at slowing or reversing the underlying atherosclerotic process by managing circulating lipids with an emphasis on control of low-density lipoprotein (LDL) cholesterol. However, over the past several decades, there has been increasing recognition that chronic inflammation and immune system activation are important contributors to atherosclerosis. This shift in focus has led to the elucidation of the complex interplay between cholesterol and cellular secretion of cytokines involved in CVD pathogenesis. Of the vast array of cytokine promoting atherosclerosis, interferon (IFN)-γ is highly implicated and, therefore, of great interest. METHODS Literature review was performed to further understand the effect of IFN-γ on the development of atherosclerotic CVD. RESULTS IFN-γ, the sole member of the type II IFN family, is produced by T cells and macrophages, and has been found to induce production of other cytokines and to have multiple effects on all stages of atherogenesis. IFN-γ activates a variety of signaling pathways, most commonly the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, to induce oxidative stress, promote foam cell accumulation, stimulate smooth muscle cell proliferation and migration into the arterial intima, enhance platelet-derived growth factor expression, and destabilize plaque. These are just a few of the contributions of IFN-γ to the initiation and progression of atherosclerotic CVD. CONCLUSION Given the pivotal role of IFN-γ in the advancement of CVD, activation of its signaling pathways is being explored as a driver of atherosclerosis. Manipulation of this key cytokine may lead to novel therapeutic avenues for CVD prevention and treatment. A number of therapies are being explored with IFN-γ as the potential target.
Collapse
Affiliation(s)
- Ailin Elyasi
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Iryna Voloshyna
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Saba Ahmed
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Lora J Kasselman
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Jennifer Behbodikhah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Allison B Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA.
| |
Collapse
|
41
|
Baardman J, Lutgens E. Regulatory T Cell Metabolism in Atherosclerosis. Metabolites 2020; 10:metabo10070279. [PMID: 32650487 PMCID: PMC7408402 DOI: 10.3390/metabo10070279] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/18/2020] [Accepted: 07/04/2020] [Indexed: 12/13/2022] Open
Abstract
Regulatory T cells (Tregs) are capable of suppressing excessive immune responses to prevent autoimmunity and chronic inflammation. Decreased numbers of Tregs and impaired suppressive function are associated with the progression of atherosclerosis, a chronic inflammatory disease of the arterial wall and the leading cause of cardiovascular disease. Therefore, therapeutic strategies to improve Treg number or function could be beneficial to preventing atherosclerotic disease development. A growing body of evidence shows that intracellular metabolism of Tregs is a key regulator of their proliferation, suppressive function, and stability. Here we evaluate the role of Tregs in atherosclerosis, their metabolic regulation, and the links between their metabolism and atherosclerosis.
Collapse
Affiliation(s)
- Jeroen Baardman
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Correspondence:
| | - Esther Lutgens
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
- Institute for Cardiovascular Prevention (IPEK), Klinikum der Universität München (KUM), Ludwig-Maximilians-Universität (LMU) München, 80336 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
42
|
Willemsen L, de Winther MPJ. Macrophage subsets in atherosclerosis as defined by single-cell technologies. J Pathol 2020; 250:705-714. [PMID: 32003464 PMCID: PMC7217201 DOI: 10.1002/path.5392] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/17/2020] [Accepted: 01/29/2020] [Indexed: 12/26/2022]
Abstract
Macrophages play a major role in the pathogenesis of atherosclerosis. Many studies have shone light on the different phenotypes and functions that macrophages can acquire upon exposure to local cues. The microenvironment of the atherosclerotic plaque contains a plethora of macrophage-controlling factors, such as cytokines, oxidised low-density lipoproteins and cell debris. Previous research has determined macrophage function within the plaque mainly by using immunohistochemistry and bulk analysis. The recent development and rapid progress of single-cell technologies, such as cytometry by time of flight and single-cell RNA sequencing, now enable comprehensive mapping of the wide range of cell types and their phenotypes present in atherosclerotic plaques. In this review we discuss recent advances applying these technologies in defining macrophage subsets residing in the atherosclerotic arterial wall of mice and men. Resulting from these studies, we describe three main macrophage subsets: resident-like, pro-inflammatory and anti-inflammatory foamy TREM2hi macrophages, which are found in both mouse and human atherosclerotic plaques. Furthermore, we discuss macrophage subset-specific markers and functions. More insights into the characteristics and phenotype of immune cells within the atherosclerotic plaque may guide future clinical approaches to treat disease. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lisa Willemsen
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Menno PJ de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Institute for Cardiovascular Prevention (IPEK)Ludwig Maximilians UniversityMunichGermany
| |
Collapse
|
43
|
Noz MP, Ter Telgte A, Wiegertjes K, Joosten LAB, Netea MG, de Leeuw FE, Riksen NP. Trained Immunity Characteristics Are Associated With Progressive Cerebral Small Vessel Disease. Stroke 2019; 49:2910-2917. [PMID: 30571420 DOI: 10.1161/strokeaha.118.023192] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background and Purpose- Cerebral small vessel disease (cSVD) is the major vascular cause of cognitive decline and dementia. The pathogenesis of cSVD remains largely unknown, although several studies suggest a role for systemic inflammation. In certain pathophysiological situations, monocytes can reprogram toward a long-term proinflammatory phenotype, which has been termed trained immunity. We hypothesize that trained immunity contributes to the progression of cSVD. Methods- Individuals with mild-to-severe cSVD participated in the study. Severity of cSVD was determined by the white matter hyperintensities (WMH) volume (mL) on magnetic resonance imaging in 2006, 2015, and the progression between 2006 and 2015 (ΔWMH). Cytokine production was assessed after ex vivo stimulation of peripheral blood mononuclear cells and monocytes. Additionally, monocyte subsets were identified by flow cytometry. Results- Fifty-one subjects (70±6 years, 60% men, 5.1±6.4 mL ΔWMH) were included. Circulating hsIL (high-sensitivity interleukin)-6 correlated with cSVD ( P=0.005, rs=0.40). Cytokine production capacity by monocytes was associated with cSVD progression. Basal IL-8 and IL-17 production ( P=0.08, rs=0.25; P=0.03, rs=0.30) and IL-6 production after Pam3Cys stimulation in monocytes was associated with cSVD (n=35: P=0.008, rs=0.44). Conversely, interferon (IFN)-γ production in Candida albicans stimulated peripheral blood mononuclear cells was negatively correlated with cSVD ( P=0.009, rs=-0.36). Flow cytometry revealed a correlation of the intermediate monocyte subset with cSVD ( P=0.01, rs=0.36). Conclusions- Severity and progression of cSVD are not only correlated with systemic inflammation (hsIL-6) but also with trained immunity characteristics of circulating monocytes, in terms of an altered cytokine production capacity and a shift toward the proinflammatory intermediate monocyte subset.
Collapse
Affiliation(s)
- Marlies P Noz
- From the Department of Internal Medicine, Radboud University Medical Center Behaviour, (M.P.N., L.A.B.J., M.G.N., N.P.R.), Nijmegen, the Netherlands
| | - Annemieke Ter Telgte
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (A.t.T., K.W., F.-E.d.L.), Nijmegen, the Netherlands
| | - Kim Wiegertjes
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (A.t.T., K.W., F.-E.d.L.), Nijmegen, the Netherlands
| | - Leo A B Joosten
- From the Department of Internal Medicine, Radboud University Medical Center Behaviour, (M.P.N., L.A.B.J., M.G.N., N.P.R.), Nijmegen, the Netherlands
| | - Mihai G Netea
- From the Department of Internal Medicine, Radboud University Medical Center Behaviour, (M.P.N., L.A.B.J., M.G.N., N.P.R.), Nijmegen, the Netherlands.,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Germany (M.G.N.)
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (A.t.T., K.W., F.-E.d.L.), Nijmegen, the Netherlands
| | - Niels P Riksen
- From the Department of Internal Medicine, Radboud University Medical Center Behaviour, (M.P.N., L.A.B.J., M.G.N., N.P.R.), Nijmegen, the Netherlands
| |
Collapse
|
44
|
Parra-Izquierdo I, Castaños-Mollor I, López J, Gómez C, San Román JA, Sánchez Crespo M, García-Rodríguez C. Lipopolysaccharide and interferon-γ team up to activate HIF-1α via STAT1 in normoxia and exhibit sex differences in human aortic valve interstitial cells. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2168-2179. [PMID: 31034990 DOI: 10.1016/j.bbadis.2019.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/17/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
In early stages of calcific aortic valve disease (CAVD), immune cells infiltrate into the valve leaflets and release cytokines such as interferon (IFN)-γ. IFN-γ has context-dependent direct effects, and also regulates other immune pathways. The purpose of this study was addressing the effects of IFN-γ on human aortic valve interstitial cells (AVICs), focusing on the pathogenic processes underlying CAVD. Strikingly, under normoxic conditions, IFN-γ induced hypoxia inducible factor (HIF)-1α expression, an effect strongly potentiated by the Toll-like receptor (TLR)-4 ligand lipopolysaccharide (LPS). Immunodetection studies confirmed the nuclear translocation of HIF-1α. Gene silencing showed that HIF-1α expression is dependent on signal transducer and activator of transcription (STAT)-1 expression. Consistent with HIF-1α induction, the secretion of the endothelial growth factor was detected by ELISA, and downregulation of the antiangiogenic factor chondromodulin-1 gene was observed by qPCR. Results also disclosed IFN-γ as a proinflammatory cytokine that cooperates with LPS to induce the expression of adhesion molecules, prostaglandin E2 and interleukins. Moreover, IFN-γ induced an osteogenic phenotype and promoted in vitro calcification that were markedly potentiated by LPS. Pharmacological experiments disclosed the involvement of Janus Kinases (JAK)/STATs as well as ERK/HIF-1α routes on the induction of calcification. Notably, IFN-γ receptor 1 expression, as well as ERK/HIF-1α activation, and the subsequent responses were more robust in male AVICs. This is the first report uncovering an immune and non-hypoxic activation of HIF-1α via STAT1 in AVIC. The aforementioned results and the sex-differential responses may be potentially relevant to better understand CAVD pathogenesis.
Collapse
Affiliation(s)
- Iván Parra-Izquierdo
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Irene Castaños-Mollor
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Javier López
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Cristina Gómez
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - J Alberto San Román
- ICICOR, Hospital Clínico Universitario, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Mariano Sánchez Crespo
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain
| | - Carmen García-Rodríguez
- Instituto de Biología y Genética Molecular, CSIC-Universidad de Valladolid, Valladolid, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
45
|
Chai JT, Ruparelia N, Goel A, Kyriakou T, Biasiolli L, Edgar L, Handa A, Farrall M, Watkins H, Choudhury RP. Differential Gene Expression in Macrophages From Human Atherosclerotic Plaques Shows Convergence on Pathways Implicated by Genome-Wide Association Study Risk Variants. Arterioscler Thromb Vasc Biol 2019; 38:2718-2730. [PMID: 30354237 PMCID: PMC6217969 DOI: 10.1161/atvbaha.118.311209] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Plaque macrophages are intricately involved in atherogenesis and plaque destabilization. We sought to identify functional pathways in human plaque macrophages that are differentially regulated in respect of (1) plaque stability and (2) lipid content. We hypothesized that differentially regulated macrophage gene sets would relate to genome-wide association study variants associated with risk of acute complications of atherosclerosis. Approach and Results— Forty patients underwent carotid magnetic resonance imaging for lipid quantification before endarterectomy. Carotid plaque macrophages were procured by laser capture microdissection from (1) lipid core and (2) cap region, in 12 recently symptomatic and 12 asymptomatic carotid plaques. Applying gene set enrichment analysis, a number of gene sets were found to selectively upregulate in symptomatic plaque macrophages, which corresponded to 7 functional pathways: inflammation, lipid metabolism, hypoxic response, cell proliferation, apoptosis, antigen presentation, and cellular energetics. Predicted upstream regulators included IL-1β, TNF-α, and NF-κB. In vivo lipid quantification by magnetic resonance imaging correlated most strongly with the upregulation of genes of the IFN/STAT1 pathways. Cross-interrogation of gene set enrichment analysis and meta-analysis gene set enrichment of variant associations showed lipid metabolism pathways, driven by genes coding for APOE and ABCA1/G1 coincided with known risk-associated SNPs (single nucleotide polymorphisms) from genome-wide association studies. Conclusions— Macrophages from recently symptomatic carotid plaques show differential regulation of functional gene pathways. There were additional quantitative relationships between plaque lipid content and key gene sets. The data show a plausible mechanism by which known genome-wide association study risk variants for atherosclerotic complications could be linked to (1) a relevant cellular process, in (2) the key cell type of atherosclerosis, in (3) a human disease-relevant setting.
Collapse
Affiliation(s)
- Joshua T Chai
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Neil Ruparelia
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Anuj Goel
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Theodosios Kyriakou
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Luca Biasiolli
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Laurienne Edgar
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Ashok Handa
- Nuffield Department of Surgical Sciences (A.H.), University of Oxford, United Kingdom
| | - Martin Farrall
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Hugh Watkins
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| | - Robin P Choudhury
- From the Division of Cardiovascular Medicine, Radcliffe Department of Medicine (J.T.C., N.R., A.G., T.K., L.B., L.E., M.F., H.W., R.P.C.), University of Oxford, United Kingdom
| |
Collapse
|
46
|
Innate Immune Dysregulation in the Development of Cardiovascular Disease in Lupus. Curr Rheumatol Rep 2019; 21:46. [DOI: 10.1007/s11926-019-0842-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Affiliation(s)
- Ziad Mallat
- From the Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, United Kingdom; and Institut National de la Santé et de la Recherche Médicale, Paris, France.
| |
Collapse
|
48
|
Getz GS, Reardon CA. Apoproteins E, A-I, and SAA in Macrophage Pathobiology Related to Atherogenesis. Front Pharmacol 2019; 10:536. [PMID: 31231209 PMCID: PMC6558525 DOI: 10.3389/fphar.2019.00536] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/29/2019] [Indexed: 01/10/2023] Open
Abstract
Macrophages are core cellular elements of both early and advanced atherosclerosis. They take up modified lipoproteins and become lipid-loaded foam cells and secrete factors that influence other cell types in the artery wall involved in atherogenesis. Apoproteins E, AI, and SAA are all found on HDL which can enter the artery wall. In addition, apoE is synthesized by macrophages. These three apoproteins can promote cholesterol efflux from lipid-loaded macrophages and have other functions that modulate macrophage biology. Mimetic peptides based on the sequence or structure of these apoproteins replicate some of these properties and are potential therapeutic agents for the treatment of atherosclerosis to reduce cardiovascular diseases.
Collapse
Affiliation(s)
- Godfrey S Getz
- Department of Pathology, The University of Chicago, Chicago, IL, United States
| | - Catherine A Reardon
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
49
|
Herder C, Roden M, Ziegler D. Novel Insights into Sensorimotor and Cardiovascular Autonomic Neuropathy from Recent-Onset Diabetes and Population-Based Cohorts. Trends Endocrinol Metab 2019; 30:286-298. [PMID: 30935671 DOI: 10.1016/j.tem.2019.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/22/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022]
Abstract
The most prevalent chronic complications of diabetes are diabetic neuropathies, among which distal sensorimotor polyneuropathy (DSPN) and cardiovascular autonomic neuropathy (CAN) are the best studied. Their major clinical sequelae such as foot ulcers, neuropathic pain, and orthostatic hypotension are associated with lower quality of life and increased risk of mortality. Here we discuss the recent insights into DSPN and CAN focusing on two prospective cohorts; that is, the German Diabetes Study (GDS) including recent-onset diabetes patients and the population-based Cooperative Health Research in the Region of Augsburg, Germany (KORA) surveys. The insights from these studies investigating novel tools for early detection and prediction of (pre)diabetic neuropathy as well as biomarkers of oxidative stress and inflammation should ultimately culminate in improving the health care of patients affected by this serious condition.
Collapse
Affiliation(s)
- Christian Herder
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; These authors contributed equally.
| | - Michael Roden
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Dan Ziegler
- Institute of Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany; German Center for Diabetes Research (DZD), Partner Düsseldorf, Düsseldorf, Germany; Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; These authors contributed equally.
| |
Collapse
|
50
|
Walker ME, Matthan NR, Solano-Aguilar G, Jang S, Lakshman S, Molokin A, Faits T, Urban JF, Johnson WE, Lamon-Fava S, Lichtenstein AH. A Western-type dietary pattern and atorvastatin induce epicardial adipose tissue interferon signaling in the Ossabaw pig. J Nutr Biochem 2019; 67:212-218. [PMID: 30981985 DOI: 10.1016/j.jnutbio.2019.02.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/17/2019] [Accepted: 02/11/2019] [Indexed: 12/17/2022]
Abstract
Epicardial adipose tissue (EAT) inflammation is thought to potentiate the development of coronary artery disease (CAD). Overall diet quality and statin therapy are important modulators of inflammation and CAD progression. Our objective was to examine the effects and interaction of dietary patterns and statin therapy on EAT gene expression in the Ossabaw pig. Pigs were randomized to 1 of 4 groups; Heart Healthy diet (high in unsaturated fat, unrefined grain, fruits/vegetables [HHD]) or Western diet (high in saturated fat, cholesterol, refined grain [WD]), with or without atorvastatin. Diets were fed in isocaloric amounts for 6 months. A two-factor edge R analysis identified the differential expression of 21 genes. Relative to the HHD, the WD resulted in a significant 12-fold increase of radical s-adenosyl methionine domain containing 2 (RSAD2), a gene induced by interferon signaling. Atorvastatin led to the significant differential expression of 17 genes predominately involved in interferon signaling. Results were similar using the Porcine Translational Research Database. Pathway analysis confirmed the up-regulation of interferon signaling in response to the WD and atorvastatin independently. An expression signature of the largely interferon related differentially expressed genes had no predictive capability on a histological assessment of atherosclerosis in the underlying coronary artery. These results suggest that a WD and atorvastatin evoke an interferon mediated immune response in EAT of the Ossabaw pig, which is not associated with the presence of atherosclerosis.
Collapse
Affiliation(s)
- Maura E Walker
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Gloria Solano-Aguilar
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Saebyeol Jang
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Sukla Lakshman
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Aleksey Molokin
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - Tyler Faits
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Joseph F Urban
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD
| | - W Evan Johnson
- Division of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, JM USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA.
| |
Collapse
|