1
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 116] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
2
|
Otunla AA, Shanmugarajah K, Davies AH, Lucia Madariaga M, Shalhoub J. The Biological Parallels Between Atherosclerosis and Cardiac Allograft Vasculopathy: Implications for Solid Organ Chronic Rejection. Cardiol Rev 2024; 32:2-11. [PMID: 38051983 DOI: 10.1097/crd.0000000000000437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Atherosclerosis and solid organ chronic rejection are pervasive chronic disease states that account for significant morbidity and mortality in developed countries. Recently, a series of shared molecular pathways have emerged, revealing biological parallels from early stages of development up to the advanced forms of pathology. These shared mechanistic processes are inflammatory in nature, reflecting the importance of inflammation in both disorders. Vascular inflammation triggers endothelial dysfunction and disease initiation through aberrant vasomotor control and shared patterns of endothelial activation. Endothelial dysfunction leads to the recruitment of immune cells and the perpetuation of the inflammatory response. This drives lesion formation through the release of key cytokines such as IFN-y, TNF-alpha, and IL-2. Continued interplay between the adaptive and innate immune response (represented by T lymphocytes and macrophages, respectively) promotes lesion instability and thrombotic complications; hallmarks of advanced disease in both atherosclerosis and solid organ chronic rejection. The aim of this study is to identify areas of overlap between atherosclerosis and chronic rejection. We then discuss new approaches to improve current understanding of the pathophysiology of both disorders, and eventually design novel therapeutics.
Collapse
Affiliation(s)
- Afolarin A Otunla
- From the Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | | | - Alun H Davies
- Section of Vascular Surgery, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Imperial Vascular Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Joseph Shalhoub
- Section of Vascular Surgery, Department of Surgery & Cancer, Imperial College London, London, United Kingdom
- Imperial Vascular Unit, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
3
|
Gong W, Tian Y, Li L. T cells in abdominal aortic aneurysm: immunomodulation and clinical application. Front Immunol 2023; 14:1240132. [PMID: 37662948 PMCID: PMC10471798 DOI: 10.3389/fimmu.2023.1240132] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration, extracellular matrix (ECM) degradation, and vascular smooth muscle cell (SMC) dysfunction. The inflammatory cells involved in AAA mainly include immune cells including macrophages, neutrophils, T-lymphocytes and B lymphocytes and endothelial cells. As the blood vessel wall expands, more and more lymphocytes infiltrate into the outer membrane. It was found that more than 50% of lymphocytes in AAA tissues were CD3+ T cells, including CD4+, CD8+T cells, γδ T cells and regulatory T cells (Tregs). Due to the important role of T cells in inflammatory response, an increasing number of researchers have paid attention to the role of T cells in AAA and dug into the relevant mechanism. Therefore, this paper focuses on reviewing the immunoregulatory role of T cells in AAA and their role in immunotherapy, seeking potential targets for immunotherapy and putting forward future research directions.
Collapse
Affiliation(s)
| | | | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
4
|
Yan Z, Zhong L, Zhu W, Chung SK, Hou P. Chinese herbal medicine for the treatment of cardiovascular diseases ─ targeting cardiac ion channels. Pharmacol Res 2023; 192:106765. [PMID: 37075871 DOI: 10.1016/j.phrs.2023.106765] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of morbidity and mortality, imposing an increasing global health burden. Cardiac ion channels (voltage-gated NaV, CaV, KVs, and others) synergistically shape the cardiac action potential (AP) and control the heartbeat. Dysfunction of these channels, due to genetic mutations, transcriptional or post-translational modifications, may disturb the AP and lead to arrhythmia, a major risk for CVD patients. Although there are five classes of anti-arrhythmic drugs available, they can have varying levels of efficacies and side effects on patients, possibly due to the complex pathogenesis of arrhythmias. As an alternative treatment option, Chinese herbal remedies have shown promise in regulating cardiac ion channels and providing anti-arrhythmic effects. In this review, we first discuss the role of cardiac ion channels in maintaining normal heart function and the pathogenesis of CVD, then summarize the classification of Chinese herbal compounds, and elaborate detailed mechanisms of their efficacy in regulating cardiac ion channels and in alleviating arrhythmia and CVD. We also address current limitations and opportunities for developing new anti-CVD drugs based on Chinese herbal medicines.
Collapse
Affiliation(s)
- Zhenzhen Yan
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Ling Zhong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China
| | - Wandi Zhu
- Cardiovascular Medicine Division and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Sookja Kim Chung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China; Faculty of Medicine & Faculty of Innovation Engineering at Macau University of Science and Technology, Taipa, Macao SAR, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
| | - Panpan Hou
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute. Zhuhai, Guangdong, China.
| |
Collapse
|
5
|
Nguyen MA, Hoang HD, Rasheed A, Duchez AC, Wyatt H, Lynn Cottee M, Graber TE, Susser L, Robichaud S, Berber İ, Geoffrion M, Ouimet M, Kazan H, Maegdefessel L, Mulvihill EE, Alain T, Rayner KJ. miR-223 Exerts Translational Control of Proatherogenic Genes in Macrophages. Circ Res 2022; 131:42-58. [PMID: 35611698 PMCID: PMC9213086 DOI: 10.1161/circresaha.121.319120] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
A significant burden of atherosclerotic disease is driven by inflammation. Recently, microRNAs (miRNAs) have emerged as important factors driving and protecting from atherosclerosis. miR-223 regulates cholesterol metabolism and inflammation via targeting both cholesterol biosynthesis pathway and NFkB signaling pathways; however, its role in atherosclerosis has not been investigated. We hypothesize that miR-223 globally regulates core inflammatory pathways in macrophages in response to inflammatory and atherogenic stimuli thus limiting the progression of atherosclerosis.
Collapse
Affiliation(s)
- My-Anh Nguyen
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Huy-Dung Hoang
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Adil Rasheed
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Anne-Claire Duchez
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Hailey Wyatt
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Mary Lynn Cottee
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Tyson E Graber
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.)
| | - Leah Susser
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Sabrina Robichaud
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - İbrahim Berber
- Electrical and Computer Engineering Graduate Program, Antalya Bilim University, Turkey (I.B.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.)
| | - Mireille Ouimet
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Hilal Kazan
- Department of Computer Engineering, Antalya Bilim University, Turkey (H.K.)
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Germany (L.M.).,Department of Medicine, Karolinska Institute, Stockholm, Sweden (L.M.)
| | - Erin E Mulvihill
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada (H.-D.H., T.E.G., T.A.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| | - Katey J Rayner
- University of Ottawa Heart Institute, Canada (M.-A.N., A.R., A.-C.D., H.W., M.L.C., L.S., S.R., M.G., M.O., E.E.M., K.J.R.).,Centre for Infection, Immunity & Inflammation, Faculty of Medicine, University of Ottawa, Canada (K.J.R.).,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Canada (M.-A.N., H.-D.H., A.R., M.L.C., L.S., S.R., M.O., E.E.M., T.A., K.J.R.)
| |
Collapse
|
6
|
Variations in the Gene Expression Profile in Atherosclerotic Patients with Non-Fatal ACS. A Preliminary Study. Int J Mol Sci 2022; 23:ijms23095017. [PMID: 35563407 PMCID: PMC9104366 DOI: 10.3390/ijms23095017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/26/2022] [Accepted: 04/29/2022] [Indexed: 11/16/2022] Open
Abstract
The pathophysiology of atherosclerosis and acute coronary syndrome (ACS) is related to interactions between immune cells, endothelium, and blood platelets. An increasing number of reports confirm the link between excessive immune activation and cellular cross-talk with ACS incidence. Our genetic and proteomic analysis was performed on strictly selected atherosclerotic patients with non-fatal ACS without typical risk factors and healthy donors. Results showed changes in the gene expression levels of the various inflammatory factors derived from the peripheral blood cells that drive the over-activation of the immune system. The enhanced activation of the immune system may lead to the overexpression of the pro-inflammatory mediators, which causes self-perpetuating machinery of processes associated with thrombosis. In our preliminary study, we confirmed an altered expression of genes associated with the inflammation and overall interaction of the vascular microenvironment. Furthermore, 5 of 92 analyzed genes, CCL2, CCR2, CSF2, GZMB, and ICOS, were expressed only in patients with ACS. In conclusion, the augmented expression of the pro-inflammatory genes from the peripheral blood cells may be a crucial genetic factor leading to the occurrence of acute inflammation and thus be significant in ACS pathogenesis.
Collapse
|
7
|
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
|
8
|
Ruiz-Limon P, Ladehesa-Pineda ML, Lopez-Medina C, Lopez-Pedrera C, Abalos-Aguilera MC, Barbarroja N, Arias-Quiros I, Perez-Sanchez C, Arias-de la Rosa I, Ortega-Castro R, Escudero-Contreras A, Collantes-Estevez E, Jimenez-Gomez Y. Potential Role and Impact of Peripheral Blood Mononuclear Cells in Radiographic Axial Spondyloarthritis-Associated Endothelial Dysfunction. Diagnostics (Basel) 2021; 11:diagnostics11061037. [PMID: 34199950 PMCID: PMC8226914 DOI: 10.3390/diagnostics11061037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Endothelial dysfunction (ED) is well known as a process that can lead to atherosclerosis and is frequently presented in radiographic axial spondyloarthritis (r-axSpA) patients. Here, we investigated cellular and molecular mechanisms underlying r-axSpA-related ED, and analyzed the potential effect of peripheral blood mononuclear cells (PBMCs) in promoting endothelial injury in r-axSpA. A total of 30 r-axSpA patients and 32 healthy donors (HDs) were evaluated. The endothelial function, inflammatory and atherogenic profile, and oxidative stress were quantified. In vitro studies were designed to evaluate the effect of PBMCs from r-axSpA patients on aberrant endothelial activation. Compared to HDs, our study found that, associated with ED and the plasma proatherogenic profile present in r-axSpA, PBMCs from these patients displayed a pro-oxidative, proinflammatory, and proatherogenic phenotype, with most molecular changes noticed in lymphocytes. Correlation studies revealed the relationship between this phenotype and the microvascular function. Additional in vitro studies confirmed that PBMCs from r-axSpA patients promoted endothelial injury. Altogether, this study suggests the relevance of r-axSpA itself as a strong and independent cardiovascular risk factor, contributing to a dysfunctional endothelium and atherogenic status by aberrant activation of PBMCs. Lymphocytes could be the main contributors in the development of ED and subsequent atherosclerosis in this pathology.
Collapse
Affiliation(s)
- Patricia Ruiz-Limon
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
- UGC of Endocrinology and Nutrition, The Biomedical Research Institute of Málaga (IBIMA), Virgen de la Victoria Hospital, 29010 Málaga, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
- Correspondence: (P.R.-L.); (Y.J.-G.)
| | - Maria L. Ladehesa-Pineda
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Clementina Lopez-Medina
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Chary Lopez-Pedrera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Maria C. Abalos-Aguilera
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Nuria Barbarroja
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Isabel Arias-Quiros
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Carlos Perez-Sanchez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Ivan Arias-de la Rosa
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Rafaela Ortega-Castro
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Alejandro Escudero-Contreras
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Eduardo Collantes-Estevez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
| | - Yolanda Jimenez-Gomez
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), 14004 Córdoba, Spain; (M.L.L.-P.); (C.L.-M.); (C.L.-P.); (M.C.A.-A.); (N.B.); (I.A.-Q.); (C.P.-S.); (I.A.-d.l.R.); (R.O.-C.); (A.E.-C.); (E.C.-E.)
- UGC Rheumatology, Reina Sofia University Hospital, 14004 Córdoba, Spain
- Department of Medicine (Medicine, Dermatology and Otorhinolaryngology), University of Córdoba, 14004 Córdoba, Spain
- Correspondence: (P.R.-L.); (Y.J.-G.)
| |
Collapse
|
9
|
Huang SC, Kao YH, Shih SF, Tsai MC, Lin CS, Chen LW, Chuang YP, Tsui PF, Ho LJ, Lai JH, Chen SJ. Epigallocatechin-3-gallate exhibits immunomodulatory effects in human primary T cells. Biochem Biophys Res Commun 2021; 550:70-76. [PMID: 33689882 DOI: 10.1016/j.bbrc.2021.02.132] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/24/2021] [Indexed: 01/07/2023]
Abstract
T cells secrete several inflammatory cytokines that play a critical role in the progression of atherosclerosis. Although green tea epigallocatechin-3-gallate (EGCG) exerts anti-inflammatory and anti-atherosclerotic effects in animals, few studies have identified the mechanism underlying these effects in human primary T cells. This study investigated the pathway involved in EGCG modulation of cytokine secretion in activated human primary T cells. We pre-treated human primary T cells with EGCG (0.1, 1, 5, 10, and 20 μM) for 4 h and incubated them with or without phorbol 12-myristate 13-acetate and ionomycin (P/I) for 20 h. The cytokine production, activator protein (AP)-1 binding activity, and level of mitogen-activated protein kinase (MAPK) were assessed using enzyme-linked immunosorbent assay, electrophoretic mobility shift assay, and Western blotting, respectively. At 10 and 20 μM, EGCG decreased interleukin (IL)-2 levels by 26.0% and 38.8%, IL-4 levels by 41.5% and 55.9%, INF-γ levels by 31.3% and 34.7%, and tumor-necrosis factor (TNF)-α levels by 23.0% and 37.6%, respectively. In addition, the level of phosphorylated c-Jun N-terminal (p-JNK) and extracellular signal-regulated kinase (p-ERK) was decreased, but not the level of p-p38 MAPK. EGCG did not alter any of the total protein amounts, suggesting a selective effect on specific types of MAPKs in stimulated human T cells. EGCG tended to inactivate AP-1 DNA-binding activity. The P/I-induced production of IL-2, IL-4, INF-γ, and TNF-α by human T cells was suppressed by AP-1 inhibitor in a concentration-dependent manner. In conclusion, EGCG suppressed cytokine secretion in activated human primary T cells, and this effect was likely mediated by AP-1 inactivation through the ERK and JNK, but not p38 MAPK, pathways. These results may be related to the mechanisms through which EGCG inhibits immune- or inflammation-related atherogenesis.
Collapse
Affiliation(s)
- Shih-Chung Huang
- Division of Cardiology, Department of Medicine, Kaohsiung Armed Forces General Hospital, Taiwan
| | - Yung-Hsi Kao
- Department of Life Sciences, National Central University, Jhongli, Taoyuan, 32001, Taiwan
| | - Shao-Fu Shih
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Liv Weichien Chen
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 11490, Taiwan
| | - Yi-Ping Chuang
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Pi-Fen Tsui
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Miaoli, Taiwan
| | - Jenn-Haung Lai
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Gueishan, Taoyuan, Taiwan
| | - Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
10
|
Truong R, Thankam FG, Agrawal DK. Immunological mechanisms underlying sterile inflammation in the pathogenesis of atherosclerosis: potential sites for intervention. Expert Rev Clin Immunol 2020; 17:37-50. [PMID: 33280442 DOI: 10.1080/1744666x.2020.1860757] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Introduction: Innate and adaptive immunity play a critical role in the underlying pathological mechanisms of atherosclerosis and potential target sites of sterile inflammation open opportunities to develop novel therapeutics. In response to oxidized LDL in the intimal layer, T cell subsets are recruited and activated at the site of atheroma to upregulate pro-atherogenic cytokines which exacerbate plaque formation instability.Areas covered: A systematic search of PubMed and the Web of Science was performed between January 2001- September 2020 and relevant articles in sterile inflammation and atherosclerosis were critically reviewed. The original information was collected on the interconnection between danger associated molecular patterns (DAMPs) as the mediators of sterile inflammation and the receptor complex of CD36-TLR4-TLR6 that primes and activates inflammasomes in the pathophysiology of atherosclerosis. Mediators of sterile inflammation are identified to target therapeutic strategies in the management of atherosclerosis.Expert opinion: Sterile inflammation via NLRP3 inflammasome is perpetuated by the activation of IL-1β and IL-18 and induction of pyroptosis resulting in the release of additional inflammatory cytokines and DAMPs. Challenges with current inhibitors of the NLRP3 inflammasome lie in the specificity, stability, and efficacy in targeting the NLRP3 inflammasome constituents without ameliorating upstream or downstream responses necessary for survival.
Collapse
Affiliation(s)
- Roland Truong
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
11
|
Munjal A, Khandia R. Atherosclerosis: orchestrating cells and biomolecules involved in its activation and inhibition. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 120:85-122. [PMID: 32085889 DOI: 10.1016/bs.apcsb.2019.11.002] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The term atherosclerosis refers to the condition of deposition of lipids and other substances in and on the artery walls, called as plaque that restricts the normal blood flow. The plaque may be stable or unstable in nature. Unstable plaque can burst and trigger clot formation adding further adversities. The process of plaque formation involves various stages including fatty streak, intermediate or fibro-fatty lesion and advanced lesion. The cells participating in the formation of atherosclerotic plaque include endothelial cells, vascular smooth muscle cells (VSMC), monocytes, monocytes derived macrophages, macrophages and dendritic cells and regulatory T cells (TREG). The role of a variety of cytokines and chemokines have been studied which either help in progression of atherosclerotic plaque or vice versa. The cytokines involved in atherosclerotic plaque formation include IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-9, IL-10, IL-12, IL-13, IL-15, IL-17, IL-18, IL-20, IL-25, IL-27, IL-33, IL-37, TNF-α, TGF-β and IFN-γ; whereas amongst the chemokines (family of small cytokines) are CCL2, CCL3, CXCL4, CCL5, CXCL1, CX3CL1, CCL17, CXCL8, CXCL10, CCL20, CCL19 and CCL21 and macrophage migration-inhibitory factor. These are involved in the atherosclerosis advancements, whereas the chemokine CXCL12 is play atheroprotective roles. Apart this, contradictory functions have been documented for few other chemokines such as CXCL16. Since the cytokines and chemokines are amongst the key molecules involved in orchestrating the atherosclerosis advancements, targeting them might be an effective strategy to encumber the atherosclerotic progression. Blockage of cytokines and chemokines via the means of broad-spectrum inhibitors, neutralizing antibodies, usage of decoy receptors or RNA interference have been proved to be useful intervention against atherosclerosis.
Collapse
Affiliation(s)
- Ashok Munjal
- Department of Genetics, Barkatullah University, Bhopal, MP, India
| | - Rekha Khandia
- Department of Genetics, Barkatullah University, Bhopal, MP, India
| |
Collapse
|
12
|
Brandsma E, Kloosterhuis NJ, Koster M, Dekker DC, Gijbels MJJ, van der Velden S, Ríos-Morales M, van Faassen MJR, Loreti MG, de Bruin A, Fu J, Kuipers F, Bakker BM, Westerterp M, de Winther MPJ, Hofker MH, van de Sluis B, Koonen DPY. A Proinflammatory Gut Microbiota Increases Systemic Inflammation and Accelerates Atherosclerosis. Circ Res 2019; 124:94-100. [PMID: 30582442 PMCID: PMC6325767 DOI: 10.1161/circresaha.118.313234] [Citation(s) in RCA: 276] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Rationale: Several studies have suggested a role for the gut microbiota in inflammation and atherogenesis. A causal relation relationship between gut microbiota, inflammation, and atherosclerosis has not been explored previously. Objective: Here, we investigated whether a proinflammatory microbiota from Caspase1−/− (Casp1−/−) mice accelerates atherogenesis in Ldlr−/− mice. Method and Results: We treated female Ldlr−/− mice with antibiotics and subsequently transplanted them with fecal microbiota from Casp1−/− mice based on a cohousing approach. Autologous transplantation of fecal microbiota of Ldlr−/− mice served as control. Mice were cohoused for 8 or 13 weeks and fed chow or high-fat cholesterol–rich diet. Fecal samples were collected, and factors related to inflammation, metabolism, intestinal health, and atherosclerotic phenotypes were measured. Unweighted Unifrac distances of 16S rDNA (ribosomal DNA) sequences confirmed the introduction of the Casp1−/− and Ldlr−/− microbiota into Ldlr−/− mice (referred to as Ldlr−/−(Casp1−/−) or Ldlr−/−(Ldlr−/−) mice). Analysis of atherosclerotic lesion size in the aortic root demonstrated a significant 29% increase in plaque size in 13-week high-fat cholesterol–fed Ldlr−/−(Casp1−/−) mice compared with Ldlr−/−(Ldlr−/−) mice. We found increased numbers of circulating monocytes and neutrophils and elevated proinflammatory cytokine levels in plasma in high-fat cholesterol–fed Ldlr−/−(Casp1−/−) compared with Ldlr−/−(Ldlr−/−) mice. Neutrophil accumulation in the aortic root of Ldlr−/−(Casp1−/−) mice was enhanced compared with Ldlr−/−(Ldlr−/−) mice. 16S-rDNA-encoding sequence analysis in feces identified a significant reduction in the short-chain fatty acid–producing taxonomies Akkermansia, Christensenellaceae, Clostridium, and Odoribacter in Ldlr−/−(Casp1−/−) mice. Consistent with these findings, cumulative concentrations of the anti-inflammatory short-chain fatty acids propionate, acetate and butyrate in the cecum were significantly reduced in 13-week high-fat cholesterol–fed Ldlr−/−(Casp1−/−) compared with Ldlr−/−(Ldlr−/−) mice. Conclusions: Introduction of the proinflammatory Casp1−/− microbiota into Ldlr−/− mice enhances systemic inflammation and accelerates atherogenesis.
Collapse
Affiliation(s)
- Eelke Brandsma
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Niels J Kloosterhuis
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Mirjam Koster
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands.,Department of Pathobiology, Dutch Molecular Pathology Center, Utrecht University, the Netherlands (M.K., A.d.B.)
| | - Daphne C Dekker
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Marion J J Gijbels
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, the Netherlands (M.J.J.G., S.v.d.V., M.P.J.d.W.).,Department of Pathology and Department of Molecular Genetics, CARIM, Maastricht University, the Netherlands (M.J.J.G.)
| | - Saskia van der Velden
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, the Netherlands (M.J.J.G., S.v.d.V., M.P.J.d.W.)
| | - Melany Ríos-Morales
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Martijn J R van Faassen
- Department of Laboratory Medicine, University Medical Center Groningen (M.J.R.v.F., F.K.), University of Groningen, the Netherlands
| | - Marco G Loreti
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Alain de Bruin
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands.,Department of Pathobiology, Dutch Molecular Pathology Center, Utrecht University, the Netherlands (M.K., A.d.B.)
| | - Jingyuan Fu
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands.,Department of Genetics (J.F.), University of Groningen, the Netherlands
| | - Folkert Kuipers
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen (M.J.R.v.F., F.K.), University of Groningen, the Netherlands
| | - Barbara M Bakker
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Marit Westerterp
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Menno P J de Winther
- Experimental Vascular Biology, Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, the Netherlands (M.J.J.G., S.v.d.V., M.P.J.d.W.).,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University, Munich, Germany (M.P.J.d.W.)
| | - Marten H Hofker
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Bart van de Sluis
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| | - Debby P Y Koonen
- From the Department of Pediatrics (E.B., N.J.K., M.K., D.C.D., M.R-M., M.G.L., A.d.B., J.F., F.K., B.M.B., M.W., M.H.H., B.v.d.S., D.P.Y.K.), University of Groningen, the Netherlands
| |
Collapse
|
13
|
Zhu YR, Jiang XX, Zhang DM. Critical regulation of atherosclerosis by the KCa3.1 channel and the retargeting of this therapeutic target in in-stent neoatherosclerosis. J Mol Med (Berl) 2019; 97:1219-1229. [DOI: 10.1007/s00109-019-01814-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 05/07/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023]
|
14
|
Arumugam P, Carroll KL, Berceli SA, Barnhill S, Wrenshall LE. Expression of a Functional IL-2 Receptor in Vascular Smooth Muscle Cells. THE JOURNAL OF IMMUNOLOGY 2018; 202:694-703. [PMID: 30598511 DOI: 10.4049/jimmunol.1701151] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/23/2018] [Indexed: 11/19/2022]
Abstract
Many nonlymphoid cell types express at least two, if not all three, subunits of the IL-2R; although, compared with lymphocytes, relatively little is known about how IL-2 affects the function of nonlymphoid cells. The limited information available suggests that IL-2 has a substantial impact on cells such as gastrointestinal epithelial cells, endothelial cells, and fibroblasts. In a previous report from our laboratory, we noted that IL-2 and IL-2Rβ-deficient mice lose smooth muscle cells over time, eventually resulting in aneurysmal aortas and ectatic esophagi. This finding, combined with our work showing that IL-2 surrounds vascular smooth muscle cells by association with perlecan, led us to ask whether vascular smooth muscle cells express an IL-2R. Toward this end, we reported the expression of IL-2Rβ on human and murine vascular smooth muscle cells. We now report that vascular smooth muscle cells express all three subunits of the IL-2R, and that expression of IL-2Rα varies with vascular smooth muscle cell phenotype. Furthermore, we show that, through a functional IL-2R, IL-2 initiates signaling pathways and impacts vascular smooth muscle cell function. Finally, we demonstrate that IL-2 expression increases upon initiation of conditions that promote intimal hyperplasia, suggesting a mechanism by which the IL-2/IL-2R system may impact this widespread vascular pathology.
Collapse
Affiliation(s)
- Prakash Arumugam
- Boonshoft School of Medicine, Wright State University, Dayton, OH 45435;
| | - Katie L Carroll
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH 45435
| | - Scott A Berceli
- Department of Surgery, University of Florida, Gainesville, FL 32611.,Malcolm Randall Veteran's Administration Medical Center, Gainesville, FL 32611; and
| | - Spencer Barnhill
- Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| | - Lucile E Wrenshall
- Department of Neuroscience, Cell Biology, and Physiology, Wright State University, Dayton, OH 45435; .,Department of Surgery, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435
| |
Collapse
|
15
|
Fernandez DM, Clemente JC, Giannarelli C. Physical Activity, Immune System, and the Microbiome in Cardiovascular Disease. Front Physiol 2018; 9:763. [PMID: 30013482 PMCID: PMC6036301 DOI: 10.3389/fphys.2018.00763] [Citation(s) in RCA: 17] [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: 03/22/2018] [Accepted: 05/30/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular health is a primary research focus, as it is a leading contributor to mortality and morbidity worldwide, and is prohibitively costly for healthcare. Atherosclerosis, the main driver of cardiovascular disease, is now recognized as an inflammatory disorder. Physical activity (PA) may have a more important role in cardiovascular health than previously expected. This review overviews the contribution of PA to cardiovascular health, the inflammatory role of atherosclerosis, and the emerging evidence of the microbiome as a regulator of inflammation.
Collapse
Affiliation(s)
- Dawn M. Fernandez
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chiara Giannarelli
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
16
|
Ray M, Autieri MV. Regulation of pro- and anti-atherogenic cytokines. Cytokine 2017; 122:154175. [PMID: 29221669 DOI: 10.1016/j.cyto.2017.09.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/29/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022]
Abstract
Despite advances in prevention and treatment, vascular diseases continue to account for significant morbidity and mortality in the developed world. Incidence is expected to worsen as the number of patients with common co-morbidities linked with atherosclerotic vascular disease, such as obesity and diabetes, continues to increase, reaching epidemic proportions. Atherosclerosis is a lipid-driven vascular inflammatory disease involving multiple cell types in various stages of inflammation, activation, apoptosis, and necrosis. One commonality among these cell types is that they are activated and communicate with each other in a paracrine fashion via a complex network of cytokines. Cytokines mediate atherogenesis by stimulating expression of numerous proteins necessary for induction of a host of cellular responses, including inflammation, extravasation, proliferation, apoptosis, and matrix production. Cytokine expression is regulated by a number of transcriptional and post-transcriptional mechanisms. In this context, proteins that control and fine-tune cytokine expression can be considered key players in development of atherosclerosis and also represent targets for rational drug therapy to combat this disease. This review will describe the cellular and molecular mechanisms that drive atherosclerotic plaque progression and present key cytokines that participate in this process. We will also describe RNA binding proteins that mediate cytokine mRNA stability and regulate cytokine abundance. Identification and characterization of the cytokines and proteins that regulate their abundance are essential to our ability to identify therapeutic approaches to ameliorate atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Mitali Ray
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Michael V Autieri
- Department of Physiology, Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
17
|
Kyaw T, Tipping P, Toh BH, Bobik A. Killer cells in atherosclerosis. Eur J Pharmacol 2017; 816:67-75. [PMID: 28483458 DOI: 10.1016/j.ejphar.2017.05.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 04/03/2017] [Accepted: 05/04/2017] [Indexed: 01/15/2023]
Abstract
Cytotoxic lymphocytes (killer cells) play a critical role in host defence mechanisms, protecting against infections and in tumour surveillance. They can also exert detrimental effects in chronic inflammatory disorders and in autoimmune diseases. Tissue cell death and necrosis are prominent features of advanced atherosclerotic lesions including vulnerable/unstable lesions which are largely responsible for most heart attacks and strokes. Evidence for accumulation of killer cells in both human and mouse lesions together with their cytotoxic potential strongly suggest that these cells contribute to cell death and necrosis in lesions leading to vulnerable plaque development and potentially plaque rupture. Killer cells can be divided into two groups, adaptive and innate immune cells depending on whether they require antigen presentation for activation. Activated killer cells detect damaged or stressed cells and kill by cytotoxic mechanisms that include perforin, granzymes, TRAIL or FasL and in some cases TNF-α. In this review, we examine current knowledge on killer cells in atherosclerosis, including CD8 T cells, CD28- CD4 T cells, natural killer cells and γδ-T cells, mechanisms responsible for their activation, their migration to developing lesions and effector functions. We also discuss pharmacological strategies to prevent their deleterious vascular effects by preventing/limiting their cytotoxic effects within atherosclerotic lesions as well as potential immunomodulatory therapies that might better target lesion-resident killer cells, to minimise any compromise of the immune system, which could result in increased susceptibility to infections and reductions in tumour surveillance.
Collapse
Affiliation(s)
- Tin Kyaw
- Baker Heart and Diabetes Institute, Melbourne, Australia; Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Australia.
| | - Peter Tipping
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Australia
| | - Ban-Hock Toh
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Melbourne, Australia
| | - Alex Bobik
- Baker Heart and Diabetes Institute, Melbourne, Australia; Department of Immunology, Monash University, Melbourne, Australia
| |
Collapse
|
18
|
Loss of ADAMTS4 reduces high fat diet-induced atherosclerosis and enhances plaque stability in ApoE(-/-) mice. Sci Rep 2016; 6:31130. [PMID: 27491335 PMCID: PMC4974561 DOI: 10.1038/srep31130] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 07/11/2016] [Indexed: 11/08/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by formation of lipid-rich plaques on the inner walls of arteries. ADAMTS4 (a disintegrin-like and metalloproteinase with thrombospondin motifs-4) is a secreted proteinase that regulates versican turnover in the arterial wall and atherosclerotic plaques. Recent reports indicated elevated ADAMTS4 level in human atherosclerotic plaques and in the plasma of acute coronary syndrome patients. Nevertheless, whether increased ADAMTS4 is a consequence of atherosclerosis or ADAMTS4 has a causal role in atherogenesis remains unknown. In this work, we investigated the role of ADAMTS4 in diet induced atherosclerosis using apolipoprotein E deficient (ApoE(-/-)) and Adamts4 knockout mice. We show that ADAMTS4 expression increases in plaques as atherosclerosis progresses in ApoE(-/-) mice. ApoE(-/-)Adamts4(-/-) double knockout mice presented a significant reduction in plaque burden at 18 weeks of age. Loss of ADAMTS4 lead to a more stable plaque phenotype with a significantly reduced plaque vulnerability index characterized by reduced lipid content and macrophages accompanied with a significant increase in smooth muscle cells, collagen deposition and fibrotic cap thickness. The reduced atherosclerosis is accompanied by an altered plasma inflammatory cytokine profile. These results demonstrate for the first time that ADAMTS4 contributes to diet induced atherosclerosis in ApoE(-/-) mice.
Collapse
|
19
|
Wasiak S, Gilham D, Tsujikawa LM, Halliday C, Norek K, Patel RG, McLure KG, Young PR, Gordon A, Kulikowski E, Johansson J, Sweeney M, Wong NC. Data on gene and protein expression changes induced by apabetalone (RVX-208) in ex vivo treated human whole blood and primary hepatocytes. Data Brief 2016; 8:1280-8. [PMID: 27570805 PMCID: PMC4990638 DOI: 10.1016/j.dib.2016.07.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 07/05/2016] [Accepted: 07/22/2016] [Indexed: 01/20/2023] Open
Abstract
Apabetalone (RVX-208) inhibits the interaction between epigenetic regulators known as bromodomain and extraterminal (BET) proteins and acetyl-lysine marks on histone tails. Data presented here supports the manuscript published in Atherosclerosis “RVX-208, a BET-inhibitor for Treating Atherosclerotic Cardiovascular Disease, Raises ApoA-I/HDL and Represses Pathways that Contribute to Cardiovascular Disease” (Gilham et al., 2016) [1]. It shows that RVX-208 and a comparator BET inhibitor (BETi) JQ1 increase mRNA expression and production of apolipoprotein A-I (ApoA-I), the main protein component of high density lipoproteins, in primary human and African green monkey hepatocytes. In addition, reported here are gene expression changes from a microarray-based analysis of human whole blood and of primary human hepatocytes treated with RVX-208.
Collapse
|
20
|
Rather RA, Malik VS, Trikha D, Bhat O, Dhawan V. Aqueous Terminalia arjuna extract modulates expression of key atherosclerosis-related proteins in a hypercholesterolemic rabbit: A proteomic-based study. Proteomics Clin Appl 2016; 10:750-759. [PMID: 26934842 DOI: 10.1002/prca.201500114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/15/2016] [Accepted: 02/29/2016] [Indexed: 07/07/2024]
Abstract
PURPOSE The present study evaluates the effect of an aqueous extract of Terminalia arjuna (aqTAE) on protein expression in aortic plaques of hypercholesterolemic rabbits using a proteomic approach. EXPERIMENTAL DESIGN Thirty male New Zealand rabbits (n = 6) were employed as Gp1 (stock diet); Gp2 (high-fat diet [HFD]); Gp3 (stock diet + aqTAE); Gp4 (HFD + aqTAE); and Gp5 (HFD + atorvastatin) and followed for 6 months. Protein lysates of aortic tissues were separated by 2DE and proteins were identified by MALDI-TOF/MS. RESULTS Serum lipids were found to be significantly increased by an HFD and reduced by aqTAE both at 3 and 6 months (Gp4 vs. Gp2; p < 0.05). Total 79 spots were differentially expressed, among which 60 individual proteins were identified, 31 grouped as atherosclerosis-related proteins and 29 classified as others. aqTAE significantly attenuated the protein expression of tumor necrosis factor α, cyclooxygenase-2, MMP-9, HSP60, ICAM-5, Endothelin-3, Vimentin, Protein S100-A9 besides others. Many of the observed proteins are known to be consistently associated with endothelial dysfunction, inflammation, plaque rupture, and immune imbalance. CONCLUSIONS AND CLINICAL RELEVANCE Strong hypolipidemic effects of aqTAE and attenuation of these signature atherogenic biomarkers using proteomics highlights the fact that aqTAE may be useful in the prevention and management of atherosclerosis.
Collapse
Affiliation(s)
- Riyaz Ahmad Rather
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Vivek Singh Malik
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Dimple Trikha
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Owais Bhat
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| | - Veena Dhawan
- Department of Experimental Medicine and Biotechnology, PGIMER, Chandigarh, India
| |
Collapse
|
21
|
Spitz C, Winkels H, Bürger C, Weber C, Lutgens E, Hansson GK, Gerdes N. Regulatory T cells in atherosclerosis: critical immune regulatory function and therapeutic potential. Cell Mol Life Sci 2016; 73:901-22. [PMID: 26518635 PMCID: PMC11108393 DOI: 10.1007/s00018-015-2080-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/30/2015] [Accepted: 10/22/2015] [Indexed: 12/14/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease that is mediated by innate and adaptive immune responses. The disease is characterized by sub-endothelial accumulation and modification of lipids in the artery wall triggering an inflammatory reaction which promotes lesion progression and eventual plaque rupture, thrombus formation, and the respective clinical sequelae such as myocardial infarction or stroke. During the past decade, T-cell-mediated immune responses, especially control of pro-inflammatory signals by regulatory T cells (Tregs), have increasingly attracted the interest of experimental and clinical researchers. By suppression of T cell proliferation and secretion of anti-inflammatory cytokines, such as interleukin-10 (IL-10) and transforming growth factor-β, Tregs exert their atheroprotective properties. Atherosclerosis-prone, hyperlipidemic mice harbor systemically less Tregs compared to wild-type mice, suggesting an imbalance of immune cells which affects local and systemic inflammatory and potentially metabolic processes leading to atherogenesis. Restoring or increasing Treg frequency and enhancing their suppressive capacity by various modulations may pose a promising approach for treating inflammatory conditions such as cardiovascular diseases. In this review, we briefly summarize the immunological basics of atherosclerosis and introduce the role and contribution of different subsets of T cells. We then discuss experimental data and current knowledge pertaining to Tregs in atherosclerosis and perspectives on manipulating the adaptive immune system to alleviate atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Charlotte Spitz
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Pettenkoferstr. 9, 80336, Munich, Germany
| | - Holger Winkels
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Pettenkoferstr. 9, 80336, Munich, Germany
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Christina Bürger
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Pettenkoferstr. 9, 80336, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Pettenkoferstr. 9, 80336, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Esther Lutgens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Pettenkoferstr. 9, 80336, Munich, Germany
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Göran K Hansson
- Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Norbert Gerdes
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Pettenkoferstr. 9, 80336, Munich, Germany.
| |
Collapse
|
22
|
Gao Q, Camous X, Lu YX, Lim ML, Larbi A, Ng TP. Novel inflammatory markers associated with cognitive performance: Singapore Longitudinal Ageing Studies. Neurobiol Aging 2015; 39:140-6. [PMID: 26923410 DOI: 10.1016/j.neurobiolaging.2015.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 10/22/2022]
Abstract
We identified and validated several novel inflammatory markers of cognitive performance in community-living older persons. An exploratory study (n = 83) correlated 177 inflammatory markers assayed by Luminex with the Mini-Mental State Examination (MMSE) and identified 8 inflammatory markers for enzyme-linked immunosorbent assay (ELISA) and correlations with MMSE, Montreal Cognitive Assessment (MoCA), and cognitive impairment in the validation study (n = 139). The validation study replicated the significant associations of soluble interleukin-2 receptor alpha chain (sIL-2Rα; p = 0.050), soluble tumor necrosis factor receptor 2 (sTNFR2; p = 0.002) and soluble glycoprotein 130 (sgp130; p = 0.026) with MMSE, and sIL-2Rα (p = 0.019) and sgp130 (p < 0.001) with MoCA. Significant trends of associations of tertiles of sgp130, sIL-2Rα, and sTNFR2 were found with cognitive impairment. Highly elevated estimates of association of high versus low tertiles were obtained for sgp130 (odds ratio [OR] = 4.24, 95% confidence interval [CI] 0.96-18.8), sIL-2Rα (OR = 3.94, 95% CI 0.83-18.7), and sTNFR2 (OR = 7.58, 95% CI 1.19-48.1). sgp130, sTNFR2, and sIL-2Rα are promising inflammatory markers of low cognitive performance for further investigation.
Collapse
Affiliation(s)
- Qi Gao
- Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xavier Camous
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Yan-Xia Lu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - May-Li Lim
- Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore
| | - Tze-Pin Ng
- Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
23
|
Abstract
Coronary artery disease (CAD) is the leading cause of death in the United States. Although CAD was formerly considered a lipid accumulation-mediated disease, it has now been clearly shown to involve an ongoing inflammatory response. Advances in basic science research have established the crucial role of inflammation in mediating all stages of CAD. Today, there is convincing evidence that multiple interrelated immune mechanisms interact with metabolic risk factors to initiate, promote, and ultimately activate lesions in the coronary arteries. This review aims to provide current evidence pertaining to the role of inflammation in the pathogenesis of CAD and discusses the impact of inflammatory markers and their modification on clinical outcomes.
Collapse
|
24
|
Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev 2015; 26:673-85. [PMID: 26005197 PMCID: PMC4671520 DOI: 10.1016/j.cytogfr.2015.04.003] [Citation(s) in RCA: 347] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/27/2015] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic inflammatory disorder of the arteries, is responsible for most deaths in westernized societies with numbers increasing at a marked rate in developing countries. The disease is initiated by the activation of the endothelium by various risk factors leading to chemokine-mediated recruitment of immune cells. The uptake of modified lipoproteins by macrophages along with defective cholesterol efflux gives rise to foam cells associated with the fatty streak in the early phase of the disease. As the disease progresses, complex fibrotic plaques are produced as a result of lysis of foam cells, migration and proliferation of vascular smooth muscle cells and continued inflammatory response. Such plaques are stabilized by the extracellular matrix produced by smooth muscle cells and destabilized by matrix metalloproteinase from macrophages. Rupture of unstable plaques and subsequent thrombosis leads to clinical complications such as myocardial infarction. Cytokines are involved in all stages of atherosclerosis and have a profound influence on the pathogenesis of this disease. This review will describe our current understanding of the roles of different cytokines in atherosclerosis together with therapeutic approaches aimed at manipulating their actions.
Collapse
|
25
|
|
26
|
Abstract
Atherosclerosis is an inflammatory disease of the vessel wall characterized by activation of the innate immune system, with macrophages as the main players, as well as the adaptive immune system, characterized by a Th1-dominant immune response. Cytokines play a major role in the initiation and regulation of inflammation. In recent years, many studies have investigated the role of these molecules in experimental models of atherosclerosis. While some cytokines such as TNF or IFNγ clearly had atherogenic effects, others such as IL-10 were found to be atheroprotective. However, studies investigating the different cytokines in experimental atherosclerosis revealed that the cytokine system is complex with both disease stage-dependent and site-specific effects. In this review, we strive to provide an overview of the main cytokines involved in atherosclerosis and to shed light on their individual role during atherogenesis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Center, L01-146.1, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
- Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilians University (LMU), Munich, Germany.
| |
Collapse
|
27
|
De Haro J, Esparza L, Bleda S, Varela C, Sanchez C, Acin F. Attenuation of early atherosclerotic lesions by immunotolerance with β2 glycoprotein I and the immunomodulatory effectors interleukin 2 and 10 in a murine model. J Vasc Surg 2014; 62:1625-31. [PMID: 25041990 DOI: 10.1016/j.jvs.2014.05.096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/24/2014] [Indexed: 11/24/2022]
Abstract
OBJECTIVE This study assessed the effect of cellular and humoral autoimmune response inhibition after immunization with β2-glycoprotein I (β2-GPI) and the effect of immunomodulation with interleukin (IL)-2 and IL-10 in the development of early atherosclerotic vascular lesion in a murine model. Atherosclerosis is increasingly considered a chronic inflammatory disease with pathogenic autoimmune processes. Regulatory T cells, and their cytokines, have been implicated in the inhibition of the development of atherosclerotic lesions and involved in the immunologic tolerance induction. METHODS Eight-week-old male C57BL6 LDL-receptor deficient (LDLR(-/-)) mice were fed a cholesterol-rich (2.8%), high-saturated-fat (82%) diet for a week and divided in five groups. The groups received the following intravenous immunizations: group I (control group): one dose of 5 μg β2-GPI; group II: 5 μg β2-GPI I and 1 μg IL-2; group III: 5 μg β2-GPI and 0.75 μg of IL-10; and group IV: 5 μg β2-GPI, 1 μg IL-2, and 0.75 μg IL-10. The aortas of the mice were assessed 8 weeks after inoculation to determine the aortic lesion size and composition in all groups. RESULTS β2-GPI immunization attenuated the early atherosclerotic lesions development compared with the control group (P = .001). Macroscopic and histologic aortic atherosclerotic lesions were significantly decreased in the IL-2 and IL-10-treated groups in β2-GPI-tolerant mice compared with the β2-GPI-tolerant group without cytokine injection (P = .001). The association of both cytokines did not provoke a major inhibition in the atherosclerosis development when compared with groups injected with the two cytokines separately. CONCLUSIONS The immunotolerance induction against β2-GPI attenuates the development of atherosclerosis lesions in an animal model, enhanced by downregulation of the cellular and humoral autoimmune response provoked by IL-2 and IL-10.
Collapse
Affiliation(s)
- Joaquin De Haro
- Angiology and Vascular Surgery Department, Getafe University Hospital, Getafe, Madrid, Spain.
| | - Leticia Esparza
- Angiology and Vascular Surgery Department, Getafe University Hospital, Getafe, Madrid, Spain
| | - Silvia Bleda
- Angiology and Vascular Surgery Department, Getafe University Hospital, Getafe, Madrid, Spain
| | - Cesar Varela
- Angiology and Vascular Surgery Department, Getafe University Hospital, Getafe, Madrid, Spain
| | - Carolina Sanchez
- Biomedical Research Centre, Getafe University Hospital, Getafe, Madrid, Spain
| | - Francisco Acin
- Angiology and Vascular Surgery Department, Getafe University Hospital, Getafe, Madrid, Spain
| |
Collapse
|
28
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
29
|
Magnusson LU, Lundqvist A, Karlsson MN, Skålén K, Levin M, Wiklund O, Borén J, Hultén LM. Arachidonate 15-lipoxygenase type B knockdown leads to reduced lipid accumulation and inflammation in atherosclerosis. PLoS One 2012; 7:e43142. [PMID: 22912809 PMCID: PMC3422220 DOI: 10.1371/journal.pone.0043142] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Accepted: 07/17/2012] [Indexed: 01/20/2023] Open
Abstract
Inflammation in the vascular wall is important for development of atherosclerosis. We have shown previously that arachidonate 15-lipoxygenase type B (ALOX15B) is more highly expressed in human atherosclerotic lesions than in healthy arteries. This enzyme oxidizes fatty acids to substances that promote local inflammation and is expressed in lipid-loaded macrophages (foam cells) present in the atherosclerotic lesions. Here, we investigated the role of ALOX15B in foam cell formation in human primary macrophages and found that silencing of human ALOX15B decreased cellular lipid accumulation as well as proinflammatory cytokine secretion from macrophages. To investigate the role of ALOX15B in promoting the development of atherosclerosis in vivo, we used lentiviral shRNA silencing and bone marrow transplantation to knockdown mouse Alox15b gene expression in LDL-receptor-deficient (Ldlr(-/-)) mice. Knockdown of mouse Alox15b in vivo decreased plaque lipid content and markers of inflammation. In summary, we have shown that ALOX15B influences progression of atherosclerosis, indicating that this enzyme has an active proatherogenic role.
Collapse
Affiliation(s)
- Lisa U Magnusson
- Sahlgrenska Center for Cardiovascular and Metabolic Research, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Dietrich T, Hucko T, Schneemann C, Neumann M, Menrad A, Willuda J, Atrott K, Stibenz D, Fleck E, Graf K, Menssen HD. Local delivery of IL-2 reduces atherosclerosis via expansion of regulatory T cells. Atherosclerosis 2011; 220:329-36. [PMID: 22062588 DOI: 10.1016/j.atherosclerosis.2011.09.050] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Revised: 09/20/2011] [Accepted: 09/27/2011] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Recent studies indicate that regulatory T cells (Tregs) attenuate murine atherosclerosis. Since interleukin (IL)-2 induces Tregs proliferation, we tested the impact of L19-IL2, a fusion antibody specific to extra-domain B of fibronectin (ED-B) containing an active human IL-2 molecule, in experimental atherosclerosis. METHODS AND RESULTS L19-IL2 or appropriate controls were given intravenously to 6 month old Western diet-fed apoE(-/-) mice on day 1, 3, and 5. Human IL-2 was detected on day 7 within atherosclerotic plaques of L19-IL2-treated mice, and magnetic resonance imaging of the plaques showed a significant adventitial gadolinium enhancement on day 7 and 13, suggesting microvascular leakage as a result of the pharmacodynamic activity of L19-IL2. Treatment with L19-IL2 significantly reduced the size of pre-established atherosclerotic plaques at the thoracic aorta (Sudan III stained area) and in the aortic root area (microscopic, morphometric analysis) on day 7 as compared to controls (L19, D1.3-IL2, NaCl) as well as compared to baseline (day 0). Tregs markers Foxp3 and CTLA4 were highly increased in plaques after L19-IL2 treatment compared to controls (p<0.01), whereas the macrophage marker Mac3 was significantly reduced (p<0.03). Co-treatment with IL-2-receptor blocking antibody PC61 abrogated L19-IL2-induced plaque reduction compared with IgG control (p<0.03). CONCLUSION L19-IL2 delivers functional IL-2 to pre-established atherosclerotic plaques of WD-fed apoE(-/-) mice resulting in significant plaque size reduction mediated by local Tregs.
Collapse
Affiliation(s)
- Thore Dietrich
- Department of Medicine-Cardiology, Deutsches Herzzentrum, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Stefanutti C, Vivenzio A, Ferraro PM, Morozzi C, Belotherkovsky D. Apheresis-inducible cytokine pattern change in severe, genetic dyslipidemias. Cytokine 2011; 56:835-41. [PMID: 21920769 DOI: 10.1016/j.cyto.2011.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 08/06/2011] [Accepted: 08/13/2011] [Indexed: 10/17/2022]
Abstract
OBJECTIVE The effects of direct adsorption of lipids LDL-apheresis (DALILDL-a) on plasma cytokines in two Homozygous and heterozygous familial hypercholesterolemic (HozFH, HtzFH) and in four HyperLp(a)lipoproteinemic [HyperLp(a)] patients, were evaluated. METHODS Plasma, macrophage inflammatory proteins 1α (MIP-1α), macrophage inflammatory proteins 1β (MIP-1β), monocyte chemoattractant protein-1 (MCP-1), RANTES (Regulated upon Activation, Normal T-cell Expressed, and Secreted), granulocyte-colony stimulating factor (GCSF), granulocyte macrophage-colony stimulating factor (GM-CSF), interleukin-1α (IL-1α), interleukin-1β (IL-1β), interleukin-2 (IL-2), interleukin-6 (IL-6), interferon-γ (IFN-γ), concentrations, were measured before and after LDL-a on three consecutive sessions for each patient. RESULTS MIP-1α was significantly reduced (P=0.05), while MIP-1β was significantly increased (P=0.05). Plasma MCP-1 was reduced, although not significantly, while RANTES was significantly increased (P=0.05). GCSF and GM-CSF were both significantly reduced (GM-CSF: P=0.05, GCSF: P=0.05, respectively). IL-1α level was significantly reduced (P=0.001). IL-1β, IL-6, and IFN-γ levels were significantly reduced in plasma after apheresis (IL-1β: P=0.001, IL-6: T1 P=0.001; T2 P=0.05, respectively, IFN-γ: P=0.001). IL-2 level in plasma was significantly reduced at T0, and T2, (P=0.001). However, IL-2 level showed a statistically significant increase at T1 (P=0.001). A significant correlation between IL-1α and IFN-γ was found: r=0.882 (P=0.001). CONCLUSIONS In this study LDL-a induced profound changes in several circulating cytokines and promoted anti-inflammatory and anti-atherogenic cytokine profile in plasma of patients with severe dyslipidemia, with pre-existing angiographically demonstrated Coronary heart disease (CHD), and aortic valvular disease (#=1) (AVD).
Collapse
Affiliation(s)
- Claudia Stefanutti
- Extracorporeal Therapeutic Techniques Unit-Immunohematology and Transfusion Medicine, Department of Molecular Medicine, University of Rome La Sapienza, Umberto I Hospital, Rome, Italy.
| | | | | | | | | |
Collapse
|
32
|
Maes M, Ruckoanich P, Chang YS, Mahanonda N, Berk M. Multiple aberrations in shared inflammatory and oxidative & nitrosative stress (IO&NS) pathways explain the co-association of depression and cardiovascular disorder (CVD), and the increased risk for CVD and due mortality in depressed patients. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:769-83. [PMID: 20561554 DOI: 10.1016/j.pnpbp.2010.06.008] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2010] [Revised: 05/24/2010] [Accepted: 06/09/2010] [Indexed: 11/19/2022]
Abstract
There is evidence that there is a bidirectional relationship between major depression and cardiovascular disorder (CVD): depressed patients are a population at risk for increased cardiac morbidity and mortality, and depression is more frequent in patients who suffer from CVD. There is also evidence that inflammatory and oxidative and nitrosative stress (IO&NS) pathways underpin the common pathophysiology of both CVD and major depression. Activation of these pathways may increase risk for both disorders and contribute to shared risk. The shared IO&NS pathways that may contribute to CVD and depression comprise the following: increased levels of pro-inflammatory cytokines, like interleukin-1β (IL-1β), IL-2, IL-6, IL-8, IL-12, tumor necrosis factor-α, and interferon-γ; T cell activation; increased acute phase proteins, like C-reactive protein, haptoglobin, fibrinogen and α1-antitrypsin; complement factors; increased LPS load through bacterial translocation and subsequent gut-derived inflammation; induction of indoleamine 2,3-dioxygenase with increased levels of tryptophan catabolites; decreased levels of antioxidants, like coenzyme Q10, zinc, vitamin E, glutathione and glutathione peroxidase; increased O&NS characterized by oxidative damage to low density lipoprotein (LDL) and phospholipid inositol, increased malondialdehyde, and damage to DNA and mitochondria; increased nitrosative stress; and decreased ω3 polyunsaturated fatty acids (PUFAs). The complex interplay between the abovementioned IO&NS pathways in depression results in pro-atherogenic effects and should be regarded as a risk factor to future clinical CVD and due mortality. We suggest that major depression should be added as a risk factor to the Charlson "comorbidity" index. It is advised that patients with (sub)chronic or recurrent major depression should routinely be assessed by serology tests to predict if they have an increased risk to cardiovascular disorders.
Collapse
|
33
|
Gómez-Guerrero C, Mallavia B, Egido J. Targeting Inflammation in Cardiovascular Diseases. Still a Neglected field? Cardiovasc Ther 2011; 30:e189-97. [DOI: 10.1111/j.1755-5922.2011.00274.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
34
|
Dunér P, To F, Berg K, Alm R, Björkbacka H, Engelbertsen D, Fredrikson GN, Nilsson J, Bengtsson E. Immune responses against aldehyde-modified laminin accelerate atherosclerosis in Apoe−/− mice. Atherosclerosis 2010; 212:457-65. [DOI: 10.1016/j.atherosclerosis.2010.07.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 07/13/2010] [Accepted: 07/15/2010] [Indexed: 10/19/2022]
|
35
|
Abstract
Conjugated linoleic acids (CLA) are a family of polyunsaturated fatty acids (PUFA), some isomers occurring naturally in beef and dairy products and others being formed as a result of bihydrogenation of vegetable oils to form margarine. Synthetic and natural sources of CLA may have beneficial effects in a range of inflammatory conditions including colitis, atherosclerosis, metabolic syndrome and rheumatoid arthritis. Most of the biological effects have been attributed to the cis9, trans11- (c9, t11-) and the trans10, cis12- (t10, c12-) isomers. Evidence suggests that c9, t11-CLA is responsible for the anti-inflammatory effect attributed to CLA while t10, t12-CLA appears to be responsible for anti-adipogenic effects. This review will focus on the effects of CLA on the inflammatory components associated with insulin resistance, atherosclerosis and Th1 mediated inflammatory disease, at a cellular, systemic and clinical level. Whist CLA may ameliorate certain aspects of the inflammatory response, particularly within cellular and animal models, the relevance of this has yet to be clarified within the context of human health.
Collapse
Affiliation(s)
- C M Reynolds
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | |
Collapse
|
36
|
To K, Agrotis A, Besra G, Bobik A, Toh BH. NKT cell subsets mediate differential proatherogenic effects in ApoE-/- mice. Arterioscler Thromb Vasc Biol 2009; 29:671-7. [PMID: 19251589 DOI: 10.1161/atvbaha.108.182592] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE NKT cells promote atherogenesis, but the subtypes responsible have not been identified. We investigated 2 major NKT cell subtypes (CD4+ and DN NKT) in ApoE-/- mice rendered NKT cell-deficient by day-3 neonatal thymectomy (3dTx). METHODS AND RESULTS Atherosclerosis development was studied in thymectomized ApoE-/- mice fed a high-fat diet with/without adoptively transferred NKT cells. We demonstrate NKT cell deficiency in thymectomized mice and markedly smaller atherosclerotic lesions. The reduction in lesion size was reversed by adoptive transfer of liver-derived NKT cells. Adoptive transfer of CD4+, but not DN NKT cells, into 3dTx ApoE-/- mice increased lesion size 2.5-fold. The differential effects were not attributable to differences in homing to developing atherosclerotic lesions. DN NKT cells expressed at least 3-fold higher levels of inhibitory Ly49 receptors (Ly49A, Ly49C/I, and Ly49G2) than CD4+ NKT cells, and lesions expressed large amounts of their MHC class I ligand. In vitro these inhibitory receptors initiated greater effects in DN NKT cells. Culture of each NKT cell subset with TAP-deficient (MHC class I-deficient) dendritic cells and alpha-GalCer led to secretion of similar amounts of proatherogenic cytokines IL-2, IFN-gamma, and TNF but, when cultured with MHC class I-positive dendritic cells, CD4+ NKT cells secreted more of these cytokines. CONCLUSIONS CD4+ NKT cells are responsible for the proatherogenic activity of NKT cells. Expression of inhibitory Ly49 receptors by the subtypes appears responsible for regulating their secretion of proatherogenic cytokines and their differential proatherogenic effects.
Collapse
Affiliation(s)
- Kelly To
- Vascular Biology & Atherosclerosis Laboratory, Baker IDI Heart & Diabetes Institute, PO Box 6492, St Kilda Road Central, Melbourne, Victoria 8008, Australia.
| | | | | | | | | |
Collapse
|
37
|
Abstract
Atherosclerosis is an inflammatory disease of the wall of large- and medium-sized arteries that is precipitated by elevated levels of low-density lipoprotein (LDL) cholesterol in the blood. Although dendritic cells (DCs) and lymphocytes are found in the adventitia of normal arteries, their number is greatly expanded and their distribution changed in human and mouse atherosclerotic arteries. Macrophages, DCs, foam cells, lymphocytes, and other inflammatory cells are found in the intimal atherosclerotic lesions. Beneath these lesions, adventitial leukocytes organize in clusters that resemble tertiary lymphoid tissues. Experimental interventions can reduce the number of available blood monocytes, from which macrophages and most DCs and foam cells are derived, and reduce atherosclerotic lesion burden without altering blood lipids. Under proatherogenic conditions, nitric oxide production from endothelial cells is reduced and the burden of reactive oxygen species (ROS) and advanced glycation end products (AGE) is increased. Incapacitating ROS-generating NADPH oxidase or the receptor for AGE (RAGE) has beneficial effects. Targeting inflammatory adhesion molecules also reduces atherosclerosis. Conversely, removing or blocking IL-10 or TGF-beta accelerates atherosclerosis. Regulatory T cells and B1 cells secreting natural antibodies are atheroprotective. This review summarizes our current understanding of inflammatory and immune mechanisms in atherosclerosis.
Collapse
Affiliation(s)
- Elena Galkina
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia 23507-1696;
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037;
| |
Collapse
|
38
|
Toyama K, Wulff H, Chandy KG, Azam P, Raman G, Saito T, Fujiwara Y, Mattson DL, Das S, Melvin JE, Pratt PF, Hatoum OA, Gutterman DD, Harder DR, Miura H. The intermediate-conductance calcium-activated potassium channel KCa3.1 contributes to atherogenesis in mice and humans. J Clin Invest 2008; 118:3025-37. [PMID: 18688283 PMCID: PMC2496961 DOI: 10.1172/jci30836] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Accepted: 06/25/2008] [Indexed: 12/13/2022] Open
Abstract
Atherosclerosis remains a major cause of death in the developed world despite the success of therapies that lower cholesterol and BP. The intermediate-conductance calcium-activated potassium channel KCa3.1 is expressed in multiple cell types implicated in atherogenesis, and pharmacological blockade of this channel inhibits VSMC and lymphocyte activation in rats and mice. We found that coronary vessels from patients with coronary artery disease expressed elevated levels of KCa3.1. In Apoe(-/-) mice, a genetic model of atherosclerosis, KCa3.1 expression was elevated in the VSMCs, macrophages, and T lymphocytes that infiltrated atherosclerotic lesions. Selective pharmacological blockade and gene silencing of KCa3.1 suppressed proliferation, migration, and oxidative stress of human VSMCs. Furthermore, VSMC proliferation and macrophage activation were reduced in KCa3.1(-/-) mice. In vivo therapy with 2 KCa3.1 blockers, TRAM-34 and clotrimazole, significantly reduced the development of atherosclerosis in aortas of Apoe(-/-) mice by suppressing VSMC proliferation and migration into plaques, decreasing infiltration of plaques by macrophages and T lymphocytes, and reducing oxidative stress. Therapeutic concentrations of TRAM-34 in mice caused no discernible toxicity after repeated dosing and did not compromise the immune response to influenza virus. These data suggest that KCa3.1 blockers represent a promising therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Kazuyoshi Toyama
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Heike Wulff
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - K. George Chandy
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Philippe Azam
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Girija Raman
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Takashi Saito
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Yoshimasa Fujiwara
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - David L. Mattson
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Satarupa Das
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - James E. Melvin
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Phillip F. Pratt
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Ossama A. Hatoum
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - David D. Gutterman
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - David R. Harder
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| | - Hiroto Miura
- Department of Medicine and Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Pharmacology, University of California, Davis, Davis, California, USA.
Department of Physiology and Biophysics, University of California, Irvine, Irvine, California, USA.
Division of Cardiology, Akita Kumiai General Hospital, Akita, Japan.
Division of Cardiology, Fujiwara Memorial Hospital, Akita, Japan.
Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Center for Oral Biology, University of Rochester, Rochester, New York, USA.
Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Surgery B, Haemek Medical Center, Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel.
Departments of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
Department of Veterans Affairs Medical Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
39
|
Fukagawa NK, Li M, Sabo-Attwood T, Timblin CR, Butnor KJ, Gagne J, Steele C, Taatjes DJ, Huber S, Mossman BT. Inhaled asbestos exacerbates atherosclerosis in apolipoprotein E-deficient mice via CD4+ T cells. ENVIRONMENTAL HEALTH PERSPECTIVES 2008; 116:1218-25. [PMID: 18795166 PMCID: PMC2535625 DOI: 10.1289/ehp.11172] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Accepted: 05/21/2008] [Indexed: 05/07/2023]
Abstract
BACKGROUND Associations between air pollution and morbidity/mortality from cardiovascular disease are recognized in epidemiologic and clinical studies, but the mechanisms by which inhaled fibers or particles mediate the exacerbation of atherosclerosis are unclear. OBJECTIVE AND METHODS To determine whether lung inflammation after inhalation of a well-characterized pathogenic particulate, chrysotile asbestos, is directly linked to exacerbation of atherosclerosis and the mechanisms involved, we exposed apolipoprotein E-deficient (ApoE(-/-)) mice and ApoE(-/-) mice crossed with CD4(-/-) mice to ambient air, NIEHS (National Institute of Environmental Health Sciences) reference sample of chrysotile asbestos, or fine titanium dioxide (TiO(2)), a nonpathogenic control particle, for 3, 9, or 30 days. RESULTS ApoE(-/-) mice exposed to inhaled asbestos fibers had approximately 3-fold larger atherosclerotic lesions than did TiO(2)-exposed ApoE(-/-) mice or asbestos-exposed ApoE(-/-)/CD4(-/-) double-knockout (DKO) mice. Lung inflammation and the magnitude of lung fibrosis assessed histologically were similar in asbestos-exposed ApoE(-/-) and DKO mice. Monocyte chemoattractant protein-1 (MCP-1) levels were increased in bronchoalveolar lavage fluid and plasma, and plasma concentrations correlated with lesion size (p < 0.04) in asbestos-exposed ApoE(-/-) mice. At 9 days, activator protein-1 (AP-1) and nuclear factor-kappaB (NF-kappaB), transcription factors linked to inflammation and found in the promoter region of the MCP-1 gene, were increased in aortas of asbestos-exposed ApoE(-/-) but not DKO mice. CONCLUSION Our findings show that the degree of lung inflammation and fibrosis does not correlate directly with cardiovascular effects of inhaled asbestos fibers and support a critical role of CD4(+) T cells in linking fiber-induced pulmonary signaling to consequent activation of AP-1- and NF-kappaB-regulated genes in atherogenesis.
Collapse
Affiliation(s)
- Naomi K Fukagawa
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont 05405, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kleemann R, Zadelaar S, Kooistra T. Cytokines and atherosclerosis: a comprehensive review of studies in mice. Cardiovasc Res 2008; 79:360-76. [PMID: 18487233 PMCID: PMC2492729 DOI: 10.1093/cvr/cvn120] [Citation(s) in RCA: 473] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the past few years, inflammation has emerged as a major driving force of atherosclerotic lesion development. It is now well-established that from early lesion to vulnerable plaque formation, numerous cellular and molecular inflammatory components participate in the disease process. The most prominent cells that invade in evolving lesions are monocyte-derived macrophages and T-lymphocytes. Both cell types produce a wide array of soluble inflammatory mediators (cytokines, chemokines) which are critically important in the initiation and perpetuation of the disease. This review summarizes the currently available information from mouse studies on the contribution of a specified group of cytokines expressed in atherosclerotic lesions, viz. interleukins (IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-10, IL-12, IL-18, IL-20) and macrophage-associated cytokines [tumour necrosis factor-α (TNF-α); macrophage migration inhibitory factor (MIF); interferon-γ (IFN-γ); colony stimulating factors G-CSF,-M-CSF,-GM-CSF) to atherogenesis. Emphasis is put on the consistency of the effects of these cytokines, i.e. inasmuch an effect depends on the experimental approach applied (overexpression/deletion, strain, gender, dietary conditions, and disease stage). An important outcome of this survey is (i) that only for a few cytokines there is sufficient consistent data allowing classifying them as typically proatherogenic (IL-1, IL-12, IL-18, MIF, IFN-γ, TNF-α, and M-CSF) or antiatherogenic (IL-10) and (ii) that some cytokines (IL-4, IL-6 and GM-CSF) can exert pro- or anti-atherogenic effects depending on the experimental conditions. This knowledge can be used for improved early detection, prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Robert Kleemann
- TNO-BioSciences, Gaubius-Laboratory, Department of Vascular and Metabolic Diseases, PO Box 2215, 2301 CE Leiden, The Netherlands.
| | | | | |
Collapse
|
41
|
Yang XF, Yin Y, Wang H. VASCULAR INFLAMMATION AND ATHEROGENESIS ARE ACTIVATED VIA RECEPTORS FOR PAMPs AND SUPPRESSED BY REGULATORY T CELLS. DRUG DISCOVERY TODAY. THERAPEUTIC STRATEGIES 2008; 5:125-142. [PMID: 19578482 PMCID: PMC2632857 DOI: 10.1016/j.ddstr.2008.11.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Despite significant advances in identifying the risk factors and elucidating atherosclerotic pathology, atherosclerosis remains the leading cause of morbidity and mortality in industrialized society. These risk factors independently or synergistically lead to chronic vascular inflammation, which is an essential requirement for the progression of atherosclerosis in patients. However, the mechanisms underlying the pathogenic link between the risk factors and atherosclerotic inflammation remain poorly defined. Significant progress has been made in two major areas, which are determination of the roles of the receptors for pathogen-associated molecular patterns (PAMPs) in initiation of vascular inflammation and atherosclerosis, and characterization of the roles of regulatory T cells in suppression of vascular inflammation and atherosclerosis. In this review, we focus on three related issues: (1) examining the recent progress in endothelial cell pathology, inflammation and their roles in atherosclerosis; (2) analyzing the roles of the receptors for pathogen-associated molecular patterns (PAMPs) in initiation of vascular inflammation and atherosclerosis; and (3) analyzing the advances in our understanding of suppression of vascular inflammation and atherosclerosis by regulatory T cells. Continuous improvement of our understanding of the risk factors involved in initiation and promotion of artherogenesis, will lead to the development of novel therapeutics for ischemic stroke and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiao-Feng Yang
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140
| | - Ying Yin
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140
| | - Hong Wang
- Department of Pharmacology and Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA 19140
| |
Collapse
|
42
|
Mullen A, Moloney F, Nugent AP, Doyle L, Cashman KD, Roche HM. Conjugated linoleic acid supplementation reduces peripheral blood mononuclear cell interleukin-2 production in healthy middle-aged males. J Nutr Biochem 2007; 18:658-66. [PMID: 17368881 DOI: 10.1016/j.jnutbio.2006.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Revised: 11/07/2006] [Accepted: 12/20/2006] [Indexed: 01/02/2023]
Abstract
Conjugated linoleic acid (CLA) refers to geometric and positional isomers of linoleic acid. Animal studies have shown that CLA modulates the immune system and suggest that it may have a therapeutic role in inflammatory disorders. This double-blind placebo-controlled intervention trial investigated the effects of CLA supplementation on indices of immunity relating to cardiovascular disease (CVD) in a cohort of healthy middle-aged male volunteers. Subjects were randomly assigned to supplement their diet with 2.2 g 50:50 isomeric blend of cis 9, trans 11 (c9, t11)-CLA and trans 10, cis 12 (t10, c12)-CLA or placebo daily for 8 weeks. Interleukin (IL) 2, IL-10 and tumour necrosis factor (TNF) alpha were measured in the supernatant of cultured unstimulated and concanavalin A (Con A)-stimulated peripheral blood mononuclear cells (PBMC) by ELISA. Serum IL-6 and plasma CRP were measured by ELISA and plasma fibrinogen by automated clotting assay. Gene expression was investigated by real-time RT-PCR. CLA supplementation significantly reduced Con A-stimulated PBMC IL-2 secretion (37.1%; P=.02). CLA supplementation had no significant effect on transcription of IL-2. CLA supplementation had no direct significant effects on PBMC TNFalpha or IL-10 secretion. Other inflammatory markers associated with CVD, including IL-6, CRP and fibrinogen, were not affected by CLA supplementation. This study showed that CLA supplementation reduced PBMC IL-2 secretion from Con A-stimulated PBMC but lacked effect on other markers of the human inflammatory response.
Collapse
Affiliation(s)
- Anne Mullen
- Nutrigenomics Research Group, Department of Clinical Medicine and Institute of Molecular Medicine, St. James's Hospital, Dublin 8, Ireland
| | | | | | | | | | | |
Collapse
|
43
|
Dyrøy E, Røst TH, Pettersen RJ, Halvorsen B, Gudbrandsen OA, Ueland T, Muna Z, Müller F, Nordrehaug JE, Aukrust P, Berge RK. Tetradecylselenoacetic acid, a PPAR ligand with antioxidant, antiinflammatory, and hypolipidemic properties. Arterioscler Thromb Vasc Biol 2006; 27:628-34. [PMID: 17185614 DOI: 10.1161/01.atv.0000255950.70774.d5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Antioxidants protect against oxidative stress and inflammation, which, in combination with hyperlipidemia, are important mediators of atherogenesis. Here we present a selenium-substituted fatty acid, tetradecylselenoacetic acid (TSA), which is hypothesized to have antioxidant, antiinflammatory, and hypolipidemic properties. METHODS AND RESULTS We show that TSA exerts antioxidant properties by delaying the onset of oxidation of human low density lipoprotein (LDL), by reducing the uptake of oxidized LDL in murine macrophages, and by increasing the mRNA level of superoxide dismutase in rat liver. TSA also showed antiinflammatory effects by suppressing the release of interleukin (IL)-2 and -4, and by increasing the release of IL-10 in human blood leukocytes. In addition, TSA decreased the plasma triacylglycerol level and increased the mitochondrial fatty acid beta-oxidation in rat liver. In pigs, TSA seemed to reduce coronary artery intimal thickening after percutaneous coronary intervention. In HepG2 cells TSA activated all peroxisome proliferator-activated receptors (PPARs) in a dose-dependent manner. CONCLUSIONS Our data suggest that TSA exert potent antioxidant, antiinflammatory, and hypolipidemic properties, potentially involving PPAR-related mechanisms. Based on these effects, it is tempting to hypothesize that TSA could be an interesting antiatherogenic approach to atherosclerotic disorders.
Collapse
Affiliation(s)
- Endre Dyrøy
- Institute of Medicine, Section of Medical Biochemistry, University of Bergen, Norway
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
There is growing evidence regarding the importance of inflammation in the pathogenesis of atherosclerosis and its ultimate progression to the clinical syndromes. Recently there has been an increasing interest in the role of helper T (Th) cells in atherosclerosis. The Th cells act with the macrophages and the dendritic cells via the various cytokines in bringing about a variety of changes thus leading to the progression of atherosclerosis. Atherosclerotic lesions have been seen to have increased expression of type 1 helper T (TH1) cells together with increased levels of the Th1 related cytokines. It is mainly the cytokines involved with Th1 functioning that seem to show a prominent effect, with the whole process centred around interferon gamma, making it seem like every pathway and the cytokines involved lead to a final common pathway of interferon gamma secretion; the increase or decrease of which dictates the progression of atherosclerosis and its final manifestation as the clinical syndromes.
Collapse
Affiliation(s)
- S G Baidya
- Institute of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, PR China.
| | | |
Collapse
|
45
|
Declercq V, Yeganeh B, Moshtaghi-Kashanian GR, Khademi H, Bahadori B, Moghadasian MH. Paradoxical effects of fenofibrate and nicotinic acid in apo E-deficient mice. J Cardiovasc Pharmacol 2006; 46:18-24. [PMID: 15965350 DOI: 10.1097/01.fjc.0000162764.12309.25] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Atherosclerosis is a complex vascular disease initiated by abnormal accumulation of plasma lipoproteins in the subendothelial space. Elevated levels of plasma triglycerides (TG) and low-density lipoprotein (LDL)-cholesterol as well as low concentrations of high-density lipoprotein (HDL) play a causal role in the development and progression of atherosclerotic lesions. We have shown that apolipoprotein E-deficient (apo E-KO) mice have elevated triglyceride levels plus diminished HDL concentrations. Drugs such as fenofibrate and nicotinic acid are well known to reduce TG and increase HDL levels in humans. In this study, we investigated the beneficial effects of fenofibrate and niacin on lipid profile and atherogenesis in apo E-KO mice and their wild-type counterparts. Animals were fed with a cholesterol-enriched diet supplemented with fenofibrate (0.1% wt/wt, n = 8) or nicotinic acid (0.5% wt/wt, n = 8) for 14 weeks. Body weights were recorded weekly, and plasma lipid profiles were determined at 4-week intervals. The hearts and aortas were collected and fixed for histologic and morphometric evaluations of atherosclerotic lesions. Fenofibrate treatment in apo E-KO mice paradoxically increased total cholesterol and TG by 65% and 44%, respectively, and decreased HDL-cholesterol levels by 35% as compared with controls. Similar effects of fenofibrate on cholesterol levels, but not on TG concentrations, were observed in C57BL/6 mice. Fenofibrate-treated mice had lower body weight as compared with controls. Niacin had no effect on body weight gain but failed to decrease TG or to increase HDL levels in either apo E-KO mice or their wild-type counterparts. Neither fenofibrate nor niacin significantly influenced atherogenesis in apo E-KO mice as compared with controls. In conclusion, this study shows that neither niacin nor fenofibrate has beneficial lipid-modifying and antiatherosclerosis activities in mice. Identification of mechanisms underlying paradoxical effects of fenofibrate on lipoprotein metabolisms in apo E-KO mice merits further investigation.
Collapse
Affiliation(s)
- Vanessa Declercq
- Department of Human Nutritional Sciences, St. Boniface Hospital Research Centre and University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Atherosclerosis is a chronic disease of the arterial wall where both innate and adaptive immunoinflammatory mechanisms are involved. Inflammation is central at all stages of atherosclerosis. It is implicated in the formation of early fatty streaks, when the endothelium is activated and expresses chemokines and adhesion molecules leading to monocyte/lymphocyte recruitment and infiltration into the subendothelium. It also acts at the onset of adverse clinical vascular events, when activated cells within the plaque secrete matrix proteases that degrade extracellular matrix proteins and weaken the fibrous cap, leading to rupture and thrombus formation. Cells involved in the atherosclerotic process secrete and are activated by soluble factors, known as cytokines. Important recent advances in the comprehension of the mechanisms of atherosclerosis provided evidence that the immunoinflammatory response in atherosclerosis is modulated by regulatory pathways, in which the two anti-inflammatory cytokines interleukin-10 and transforming growth factor-β play a critical role. The purpose of this review is to bring together the current information concerning the role of cytokines in the development, progression, and complications of atherosclerosis. Specific emphasis is placed on the contribution of pro- and anti-inflammatory cytokines to pathogenic (innate and adaptive) and regulatory immunity in the context of atherosclerosis. Based on our current knowledge of the role of cytokines in atherosclerosis, we propose some novel therapeutic strategies to combat this disease. In addition, we discuss the potential of circulating cytokine levels as biomarkers of coronary artery disease.
Collapse
Affiliation(s)
- Alain Tedgui
- Institut National de la Santé et de la Recherche Médicale U. 689, Cardiovascular Research Center Lariboisiere, and University Paris 7, Paris, France.
| | | |
Collapse
|
47
|
Ge J, Jia Q, Liang C, Luo Y, Huang D, Sun A, Wang K, Zou Y, Chen H. Advanced glycosylation end products might promote atherosclerosis through inducing the immune maturation of dendritic cells. Arterioscler Thromb Vasc Biol 2005; 25:2157-63. [PMID: 16100036 DOI: 10.1161/01.atv.0000181744.58265.63] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Both advanced glycosylation end products (AGEs) and dendritic cells (DCs) have been shown to play a causative role in atherosclerosis. However, whether they function interactively in the process remains uncertain. We therefore studied the effects of AGE-bovine serum albumin (AGE-BSA) on the maturation of DCs and the expressions of scavenger receptor-A (SR-A) and receptor for AGEs (RAGE) on DCs. METHODS AND RESULTS AGE-BSA induced DCs maturation accompanied with increased expressions of CD1a, CD40, CD80, CD83, CD86, and MHC class II. The capacity of DCs to stimulate T-cell proliferation and secretion of cytokines (interferon [IFN], IFN-gamma, interleukin [IL]-10 and IL-12) was also enhanced by AGE-BSA. AGE-BSA significantly upregulated SR-A and RAGE expression on DCs and the upregulation was abolished by inhibition of mitogen-activated protein (MAP) kinase Jnk, but not by that of Erk and p38 MAP kinase. AGE-BSA-induced expression of CD83 and secretion of IL-12 were partly inhibited by either an anti-RAGE neutralizing antibody or a Jnk inhibitor. CONCLUSIONS AGE-BSA induces maturation of DCs and augmented their capacity to stimulate T-cell proliferation and cytokine secretions possibly through upregulation of RAGE and SR-A, which at least in part through Jnk. These findings might explain in part the interactive roles of AGEs and DCs in the processes of atherosclerosis.
Collapse
MESH Headings
- Anthracenes/pharmacology
- Antibodies/pharmacology
- Antigens, CD/metabolism
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Cell Communication/drug effects
- Cell Communication/immunology
- Cell Differentiation/drug effects
- Cell Differentiation/immunology
- Dendritic Cells/cytology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Gene Expression/drug effects
- Gene Expression/immunology
- Glycation End Products, Advanced/pharmacology
- Humans
- Immunoglobulins/metabolism
- In Vitro Techniques
- Interleukin-12/metabolism
- JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors
- JNK Mitogen-Activated Protein Kinases/metabolism
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/immunology
- Membrane Glycoproteins/metabolism
- Monocytes/cytology
- Receptor for Advanced Glycation End Products
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Scavenger Receptors, Class A/genetics
- Scavenger Receptors, Class A/metabolism
- Serum Albumin, Bovine/pharmacology
- T-Lymphocytes/cytology
- Up-Regulation/drug effects
- Up-Regulation/immunology
- CD83 Antigen
Collapse
Affiliation(s)
- Junbo Ge
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, 180 Fenglin Rd, Shanghai 200032, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|